Next Article in Journal
Surface Modification of Polylactic Acid Bioscaffold Fabricated via 3D Printing for Craniofacial Bone Tissue Engineering
Next Article in Special Issue
Molecular Insights into Female Hybrid Sterility in Interspecific Crosses between Drosophila melanogaster and Drosophila simulans
Previous Article in Journal
Macrophage Polarization: Learning to Manage It 2.0
Previous Article in Special Issue
Expression Analysis Reveals Differentially Expressed Genes in BPH and WBPH Associated with Resistance in Rice RILs Derived from a Cross between RP2068 and TN1
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Drosophila melanogaster Transcriptome Response to Different Wolbachia Strains

by
Nataly E. Gruntenko
*,
Maksim A. Deryuzhenko
,
Olga V. Andreenkova
,
Olga D. Shishkina
,
Margarita A. Bobrovskikh
,
Natalja V. Shatskaya
and
Gennady V. Vasiliev
Institute of Cytology and Genetics SB RAS, 630090 Novosibirsk, Russia
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(24), 17411; https://doi.org/10.3390/ijms242417411
Submission received: 31 October 2023 / Revised: 26 November 2023 / Accepted: 7 December 2023 / Published: 12 December 2023
(This article belongs to the Special Issue Transcriptomics in the Study of Insect Biology)

Abstract

:
Wolbachia is a maternally inherited, intercellular bacterial symbiont of insects and some other invertebrates. Here, we investigated the effect of two different Wolbachia strains, differing in a large chromosomal inversion, on the differential expression of genes in D. melanogaster females. We revealed significant changes in the transcriptome of the infected flies compared to the uninfected ones, as well as in the transcriptome of flies infected with the Wolbachia strain, wMelPlus, compared to flies infected with the wMelCS112 strain. We linked differentially expressed genes (DEGs) from two pairwise comparisons, “uninfected—wMelPlus-infected” and “uninfected—wMelCS112-infected”, into two gene networks, in which the following functional groups were designated: “Proteolysis”, “Carbohydrate transport and metabolism”, “Oxidation–reduction process”, “Embryogenesis”, “Transmembrane transport”, “Response to stress” and “Alkaline phosphatases”. Our data emphasized similarities and differences between infections by different strains under study: a wMelPlus infection results in more than double the number of upregulated DEGs and half the number of downregulated DEGs compared to a wMelCS112 infection. Thus, we demonstrated that Wolbachia made a significant contribution to differential expression of host genes and that the bacterial genotype plays a vital role in establishing the character of this contribution.

1. Introduction

Wolbachia are widely perceived as an insect parasite since it is found in up to 40–60% of arthropod species [1,2]. However, despite the strong negative influence on the hosts shown for certain Wolbachia strains [3,4,5,6], insects have to coexist with this bacterium. It seems that there is a more ambiguous relationship between Wolbachia and their host. In some (but not all) host species, a Wolbachia infection causes a harmful outcome through four main mechanisms: cytoplasmic incompatibility (CI), male killing, feminization and parthenogenesis [7]. This might result in reduced fertility, among other things. At first glance, it is not obvious how reduced fertility might be favorable for Wolbachia. However, through these manipulations, Wolbachia are able to increase their infection rates in the host population.
As opposed to their negative influence, many of the positive effects of Wolbachia on the host have only become known in the last fifteen years [8]. Cases of vitamin supplement, antiviral protection of the host by the Wolbachia symbiont, protection from some types of stresses and even an increase in host longevity have been demonstrated [8]. In short, Wolbachia can impact their host in many ways. It is possible that the beneficial effects on survival provided by Wolbachia almost balance out the negative effects associated with reproduction.
Relationships in the Wolbachia–host symbiotic system can be described as an intricate codependency and are a result of prolonged coevolution. Wolbachia genomes are incredibly diverse since they have accumulated a vast array of differences during evolution in various hosts. Isolation is a key factor in creating such diversity. As for the hosts, the connection between their genetic diversity and the presence of Wolbachia remains to be investigated. Since our study is focused on the effect of Wolbachia on Drosophila melanogaster, we will further describe the diversity of Wolbachia genotypes in this host in particular. The widely used classification of genotypes in Wolbachia infecting D. melanogaster was suggested by Riegler et al. [9] based on a number of genome structural variants, including genome rearrangements, copy number variants and IS5 transposable elements. Later, it was improved and now includes the following Wolbachia genotypes, which are divided into two groups: wMel, wMel2, wMel3 and wMel4 (the wMel group) and wMelCS and wMelCS2 (the wMelCS group).
Different Wolbachia strains were shown to have different effects on their host. For example, Wolbachia strains transferred to the same genetic background of D. simulans from different Drosophila species provided antiviral protection with different intensities [10]. Various Wolbachia genotypes transferred to the same genetic background of D. melanogaster have also demonstrated different effects on host hormonal status and survival under heat stress [11,12]. The effects on the host depend not only on the strain of Wolbachia but also on the host’s nuclear background [12,13,14]. Evidence suggests that survival depends on strong interactions between the Wolbachia infection and host genotype and also on the mating conditions.
We conducted a detailed investigation of both adverse and beneficial effects of Wolbachia on their host. Previously, we created a conplastic line in D. melanogaster with a nuclear genotype from the wild-type line, Bi90, and the cytoplasm of the line infected with the wMelCS genotype of Wolbachia. To obtain a line carrying the same nuclear genotype but a different strain of Wolbachia, cytoplasmic substitution was achieved after 20 generations of individual saturating crosses of females carrying cytoplasm with the Wolbachia strain of interest with males from the Bi90T line. The Bi90T line was obtained from the Bi90 wild-type D. melanogaster line treated with tetracycline for three generations prior to the start of crosses. Thus, the original infection in the Bi90 line was completely eliminated, and a new line infected with the wMelCS strain, Bi90wMelCS, was obtained. This model allowed us to discover the effect of the wMelCS genotype on the overall fitness and metabolism of juvenile hormones and dopamine [11,12].
Following these discoveries, we asked ourselves if the characteristics discovered in the Bi90wMelCS line are distinctive from those of the lines infected with other isolates from the wMelCS genotype. Or have we stumbled upon a new Wolbachia strain with unique characteristics, similar to the previously described pathological strain, wMelPop [3], the difference being that our strain has a positive impact on the host? In order to answer this question, we created another model using carryover of the cytoplasm carrying Wolbachia with the wMelCS genotype from various isolates found in nature (the genotype was defined by the classification from Riegler et al. [9]) to the genetic background of the D. melanogaster line, Bi90T. Experiments on this new model showed that the increase in dopamine metabolism, body weight and lipid and glucose content is characteristic of flies infected with all strains under study, which belonged to the wMelCS genotype of Wolbachia [15,16]; however, only one line, carrying a strain of Wolbachia from line w153, was shown to increase the host’s resistance to heat stress [15]. Notably, this effect was also observed when the Wolbachia strain under study was transferred to the D. melanogaster wild-type line, Canton S [15]. The unique strain found was named wMelPlus.
We hypothesized that changes in the Wolbachia genome might cause the emergence of a stress-resistant phenotype in wMelPlus-infected flies. To better understand the underlying mechanisms of wMelPlus-induced host stress resistance, we performed whole-genome sequencing of the wMelPlus strain, which confirmed that it is, in fact, a new strain by revealing a large inversion that had not been described before [17]. The wMelCS112 and wMelPlus strains were found to share a very close genetic composition differing only by the above-mentioned chromosome inversion and by nine SNPs (seven in coding sequences, including synonymous and missense variants, and two in noncoding sequences). However, no genes that could explain the wMelPlus effect on the host were found among those altered by these SNPs. On the other hand, inversions are known to be able to strongly affect the bacterial phenotype, including colony morphology, antibiotic susceptibility, hemolytic activity and the expression of many genes [18]. The inversion in the wMelPlus genome included the well-known Octomom region, containing eight Wolbachia genes, the loss or amplification of which causes wMelPop over-proliferation [19,20]. However, only one copy of Octomom is present in the wMelPlus genome, just as in the wMelCS112 genome. Among the genes encoding products that excrete into the environment of the bacterial cell and, thereby, might be involved in the host–Wolbachia interactions [21], there are the protein-encoding gene, WMELPLUS_00535, and three copies of genes coding WsnRNA46—a long noncoding RNA in the inversion, unique for wMelPlus [17]. WMELPLUS_00535 presumably codes a transmembrane protein containing a type IV secretion system domain (TrbC/VIRB2 pilin). WsnRNA46 is known for employing the mechanism of RNA interference to upregulate a microtubular motor protein of the host, Dhc, which is important for Wolbachia transmission into Drosophila oocytes [22,23]. Though these genes might present some interest for disentangling the conundrum of the bacteria–host interaction, our main hypothesis is currently that this inversion causes dysregulation of the involved genes, which results in the disruption of the established genetic regulatory circuits and subsequent changes in Drosophila gene expression.
We can say that one inversion in a small genome is one giant leap for its host. However, we decided not to limit our examination of this symbiotic influence to the bacterium alone. After the whole genome of wMelPlus was obtained and analyzed, the next logical step was to obtain the transcriptome of flies infected with it.
The aim of our current work was to reveal the molecular mechanisms underlying the changes caused by wMelPlus Wolbachia in the host. Moreover, we searched for the differences in the changes in differential expression of genes in wMelPlus-infected flies and those infected with another strain, wMelCS112, which does not differ from other known variants of the wMelCS genotype, as it lacks the inversion that sets wMelPlus apart.
With this goal in mind, we decided to conduct transcriptomic analysis of three D. melanogaster lines: Bi90wMelPlus and Bi90wMelCS112, carrying the wMelPlus and wMelCS112 strains of Wolbachia, respectively, and the uninfected Bi90T line. It is of interest to compare the transcriptional response to the Wolbachia infection in the context of the interstrain variability.
At first, we considered using only the ovaries of flies, since it is known that these organs are rich in Wolbachia. However, it had been recently demonstrated that the effects of Wolbachia on the composition of mRNA transcripts of ovaries and early embryos of D. melanogaster are limited and variable [24,25]. The data obtained allowed Frantz et al. [25] to conclude that a Wolbachia infection explains only a small share of ovarian transcriptional variation compared with the variation among host lines. Furthermore, Wolbachia may affect genes that are expressed in other organs but not in the ovaries. It is also necessary to take into account batch effects known to be a huge contributing factor for fluctuations in the transcriptome caused by changing conditions [26]. Based on the aforementioned studies, we decided to investigate the whole female fly transcriptome, keeping in mind the importance of synchronizing insects used for the analysis in order to avoid transcriptional variation caused by factors other than the infection.

2. Results

2.1. Mapping and Quantification

Samples taken into analysis came from three Bi90T-based lines of flies: uninfected, infected with wMelPlus and infected with wMelCS112 Wolbachia strains. Four replicates for the uninfected line and three for each infected line were used to obtain pooled RNA for sequencing. Each replicate came from 20 whole, 10-day-old females. As a result, 20 RNA-seq libraries (forward and reverse reads) of 20 million reads were obtained with a length range of 75 to 76 bp. After subsequent quality control, 15.5 to 29.8 million reads per sample remained. Mapping results are presented in Table 1. Quantification was performed using featureCounts [27].

2.2. Analysis of Differentially Expressed Genes

Comparative analysis of the transcriptomes of uninfected flies and flies infected with two Wolbachia strains, wMelPlus and wMelCS112, showed significant differences in the expression levels of genes. The differences between the groups are complex and require detailed description, which is given below.
A total of 12,550 genes were identified and annotated. A total of 714 differentially expressed genes (DEGs) were found, including 493 DEGs from the comparison of the “Uninfected” and “Infected with wMelCS112” transcriptomes and 336 DEGs from the comparison of the “Uninfected” and “Infected with wMelPlus” transcriptomes. A total of 115 DEGs fell into both of these groups, which means they differentiate the Wolbachia-infected lines under study from the uninfected line.
The estimated data distribution in the principal component analysis (PCA) matched the predicted one (Figure 1). The first and second components described 77.3% of the variants. The uninfected Bi90T line differed from the infected lines by PC2; the wMelPlus-infected line differed from the two other lines under study by PC1.
For the DEGs found, gene networks were constructed using the STRING database. For the first pair, 368 nodes and 831 edges were obtained; for the second pair, 236 nodes and 615 edges were obtained. Based on the DEGs annotation, the following functional groups were designated: “Proteolysis”, “Carbohydrate transport and metabolism”, “Oxidation–reduction process”, “Embryogenesis”, “Transmembrane transport”, “Response to stress” and “Alkaline phosphatases” (Figure 2 and Figure 3).
For both comparisons made, similar functional groups of DEGs were identified, but they contained different sets of genes, as can be seen from the heat maps constructed for them (Figure 4, Figure 5, Figure 6, Figure 7, Figure 8 and Figure 9 and Figure S1). It is important to emphasize that not all genes were included in the networks obtained: only 368 DEGs from the 493 DEGs in the comparison of the “Uninfected” and “Infected with wMelCS112” transcriptomes and 236 DEGs from the 336 DEGs in the comparison of the “Uninfected” and “Infected with wMelPlus” transcriptomes are connected to each other. However, the structure of the network is highly interconnected: a total of 831 edges are included for the “Uninfected–infected with wMelCS112” comparison and a total of 615 for the “Uninfected–infected with wMelPlus” comparison. The “Transmembrane transport”, “Carbohydrate transport and metabolism”, “Oxidation–reduction process” and “Embryogenesis” functional groups turned out to be strongly connected to the “Proteolysis” group. “Proteolysis” is the largest identified group, and “Alkaline phosphatase” is the smallest one. However, both of them strongly affect the host’s organism and vital functions.
We constructed heat maps for each designated functional group: “Proteolysis” (Figure S1), “Carbohydrate transport and metabolism” (Figure 4), “Oxidation–reduction process” (Figure 5), “Embryogenesis” (Figure 6), “Transmembrane transport” (Figure 7) and “Response to stress” (Figure 8). Each functional group contains both a set of genes that distinguish flies infected with either wMelCS112 or wMelPlus from the uninfected control and genes that distinguish flies infected with wMelCS112 from those infected with wMelPlus. Samples of the same fly lines show some variation in gene expression levels, but general tendencies toward an increase or decrease in expression in infected lines relative to the uninfected control remain. In the “Carbohydrate transport and metabolism” functional group, there are more genes with higher expression levels in wMelCS112-infected flies than in wMelPlus-infected ones. However, in all the other functional groups, the opposite tendency can be seen.
The same functional groups are identified in the data of RNA-seq of samples from another batch (Table S1, Figures S2 and S3) demonstrating the reproducibility of the results obtained on the first batch.
The analysis of DEGs involved in stress response revealed similar changes in the network related to two gene families, Turandot and Bomanins, in both infected lines compared to the uninfected one (Figure 8). What is unique for wMelPlus-infected females in relation to other groups under study is the increased level of expression of the corazonin receptor gene, CrzR.
One more functional group we distinguished in our analysis is the family of alkaline phosphatase genes. It could be seen that two representatives of this family, the phu and Alp4 genes, are upregulated in females infected with the wMelPlus Wolbachia strain; and three others, the Alp2, Alp9 and Alp10 genes, are upregulated in females infected with the wMelCS112 strain (Figure 9).
Additionally, we evaluated relative expression levels of DEGs included in the functional groups. In total, 350 genes were identified as elements of these groups (Figure 10). A total of 218 genes had increased and 122 had decreased expression levels in the line infected with the wMelPlus strain relative to the uninfected control group. At the same time, a total of 94 genes had increased and 256 had decreased expression levels in the line infected with the wMelCS112 strain relative to the uninfected control group. In both infected lines, 192 genes had similar changes in the patterns of expression. More specifically, 79 genes were upregulated in both strains, while 113 were downregulated compared to the uninfected line. On the other hand, 143 genes changed their expression pattern. Seven genes increased their expression in flies infected with wMelCS112 and were decreased in flies infected with wMelPlus. At the same time, 136 genes increased their expression in the presence of wMelPlus and decreased their expression in the presence of wMelCS112.
Table 2, Table 3 and Table 4 show the distribution of annotated genes by major functional groups and the type of regulation. The tables also include those genes that are not represented in gene networks but are present in the heat maps. A clear distinction between the three lines under study can be seen. We would like to emphasize the existence of genes regulated similarly in both infected lines in all functional groups. These genes represent an overall response to the Wolbachia infection of the wMelCS group in D. melanogaster females.
Other genes in the functional groups not only differ in expression levels but also distinguish the “Uninfected—wMelCS112” pair from “Uninfected—wMelPlus”. All of the above indicates the importance of the selected functional groups.

2.3. qRT-PCR Analysis of Five Differentially Expressed Genes

We validated the RNA-seq data using qRT-PCR in five DEGs: TotA, TotC, Ugt35C1, CrzR and Lectin-33A (Figure 11). The directionality and relative magnitude of change matched the transcriptome data in all cases. The expression of TotA, TotC, Lectin-33A and Ugt35C1 was decreased in both infected strains (p < 0.001 for TotA and TotC in both Bi90wMelCS112 and Bi90wMelPlus; p < 0.01 for Ugt35C1 in Bi90wMelPlus and Lectin-33A in Bi90wMelCS112; p < 0.05 for Lectin-33A in Bi90wMelPlus). The expression level of CrzR in Bi90wMelCS112 did not differ from that in the uninfected Bi90T line, but in Bi90wMelPlus, it was significantly higher than in Bi90T (p < 0.001).

3. Discussion

Our findings showed that Wolbachia affects many biological processes in D. melanogaster females, for which the following functional groups can be designated: “Proteolysis”, “Carbohydrate transport and metabolism”, “Oxidation–reduction process”, “Embryogenesis”, “Transmembrane transport”, “Response to stress” and “Alkaline phosphatases”. Earlier, transcriptome analysis in D. melanogaster infected with Wolbachia was performed using virgin and mated females [28,29], embryos [24] and in dissected ovaries [25,30] and testes [29]. No significant differences were found in the mRNA composition between Wolbachia-infected embryos and uninfected ones [24], and the effect of Wolbachia on ovaries was rather limited: only 26 DEGs were identified in two batches 2 months apart [25]. It seems that most of the changes caused by Wolbachia in host expression start during later developmental stages. However, several gene ontology (GO) terms were found in another transcriptome analysis of Wolbachia-infected ovaries [30], including metabolic process, development process, and response to stimulus, which corresponds to our data on changes in the differential expression in the “Carbohydrate transport and metabolism” and “Embryogenesis” functional groups in Bi90wMelPlus and Bi90wMelCS112 flies compared with the uninfected Bi90T ones (see Figure 2 and Figure 3, Figures S2 and S3).
The results obtained in D. melanogaster females allowed the authors to relate them to the GO terms, iron ion binding and oxidation–reduction process [29], or create protein–protein interaction networks in STRING with the strongest interactions including “stress”, “ubiquitin”, “RNA binding and processing”, “transcription and translation” and “metabolism pathways” [28]. When comparing these results with our data, both similarities and differences were found. We also identified the “Oxidation–reduction process”, “Carbohydrate transport and metabolism” and “Response to stress” functional groups but did not find any groups connected with RNA synthesis and processing, protein synthesis or post-translational modification in the first batch (see Figure 2 and Figure 3); however, in the second batch, the “Ribosomal genes” functional group was revealed in wMelPlus-infected flies compared to the uninfected ones (see Figure S2). It seems that this functional group is rather unstable in its manifestations, which might be influenced by additional factors.
It Is worth noting that previous works on transcriptomic analysis of D. melanogaster lines carrying Wolbachia primarily focused on the wMel or wMel2 genotype [24,28,29,30]. To our knowledge, our research group was the first to conduct transcriptomic analysis of D. melanogaster lines infected with Wolbachia with a genotype from the wMelCS group. Since different Wolbachia strains were found to have different effects on the expression patterns of the host genes (see Figure 4, Table 2, Table 3 and Table 4), it could be supposed that the effect of the strains belonging to the wMel group of genotypes differed from that of the strains belonging to the wMelCS group. It is interesting that both strains having the most prominent effect on flies’ survival, wMelPop and wMelPlus, were found to be representatives of the wMelCS genotype [31], although the wMel genotype is much more widespread in nature compared to wMelCS [9,31,32,33]. The pathogenic wMelPop strain causes premature death of the D. melanogaster host [3,34,35], whereas infection with the wMelPlus strain promotes host survival under acute heat stress [11,15].
As to the results obtained in males or their testes, surprisingly, they also partially agree with our findings in female flies. Carbohydrate and lipid metabolism have been shown to be affected by Wolbachia infection both in adult testes [36] and whole males of D. melanogaster [37]. In testes, Wolbachia also affected differentially expressed genes involved in proteolysis [36], just as in Bi90wMelPlus and Bi90wMelCS112 flies (see Figure 3 and Figure 4 and Figure S1). Notably, two genes responsible for the intracellular cholesterol transport (Npc2f, Npc2d) were upregulated in Wolbachia-infected testes [36], while they were downregulated in Bi90wMelPlus and Bi90wMelCS112 females (see Figure S1). These contrasting results can probably be explained by sexual dimorphism: cholesterol is a precursor of 20-hydroxyecdysone, which, in female Drosophila, plays a major regulatory role in the control of oogenesis [38].
The following observations were also important to take into account. Both a transcriptional response to infection by any strain of Wolbachia and interstrain variability were observed simultaneously. The genotype of the symbiont largely determined the effect it had on the host insect (see Figure 4, Table 2, Table 3 and Table 4). Consequently, we can only assess a general response to the Wolbachia infection by comparing our results to other findings.
However, changes in metabolism could be considered common for Wolbachia-infected flies because they were demonstrated in several studies besides the present one [28,30,36,37]. This especially applies to carbohydrate transport and metabolism, the changes in which (see Figure 4, Table 2) correspond to increased glucose content in flies infected with different Wolbachia genotypes [16,36] (Figure 12).
It was shown earlier that Wolbachia depend on intermediate carbohydrates of the host to produce ATP and are able to manipulate the host’s energy metabolism in order to obtain these molecules [39]. In D. melanogaster, it was found that not only do infected flies have increased levels of glucose [16,36], but Wolbachia change the protein/carbohydrate ratio throughout the fly’s life [40]. This phenomenon can probably be explained by a competition for carbohydrates between Wolbachia and the host, which results in a decrease in the lifespan of infected flies under carbohydrate deficiency conditions.
The changes in proteolysis could also be considered common for flies infected with various Wolbachia strains, corresponding to changes in food consumption in the D. melanogaster lines infected with different Wolbachia genotypes and to increased starvation resistance demonstrated for the line infected with the wMelPlus strain [16,36]. It should be noted that the direction of these changes depends on sex: infected females demonstrate decreased appetite compared with uninfected ones [16], whereas infected males show increased food consumption [37]. Notably, changes in the activities of acid proteases, lipases and esterases were also found in Wolbachia-infected larvae of Habrobracon hebetor compared to uninfected ones [41].
Figure 12. Scheme demonstrating the correlation between the changes in differential expression and the previously shown metabolic and fitness changes caused by Wolbachia [7,8,16]. Green region–changes caused by any Wolbachia strain, yellow region—changes caused by wMelPlus only. TAG—triglycerides. Upward pointing arrows indicate the increase of a parameter shown in Wolbachia-infected flies compared to uninfected ones, the downward pointing arrow indicates the decrease of a parameter shown in Wolbachia-infected flies compared to uninfected ones. The question marks indicate possible connection of changes in DEGs and Wolbachia effect on the host.
Figure 12. Scheme demonstrating the correlation between the changes in differential expression and the previously shown metabolic and fitness changes caused by Wolbachia [7,8,16]. Green region–changes caused by any Wolbachia strain, yellow region—changes caused by wMelPlus only. TAG—triglycerides. Upward pointing arrows indicate the increase of a parameter shown in Wolbachia-infected flies compared to uninfected ones, the downward pointing arrow indicates the decrease of a parameter shown in Wolbachia-infected flies compared to uninfected ones. The question marks indicate possible connection of changes in DEGs and Wolbachia effect on the host.
Ijms 24 17411 g012
As mentioned above, various Wolbachia strains altered gene expression in different ways (see Figure 10): the wMelCS112 strain mostly caused a decrease in the expression level of the genes belonging to the functional groups we identified in the present study, whereas wMelPlus mostly caused an increase in their expression. A total of 256 genes were downregulated in the wMelCS112-infected flies and only 122 in the wMelPlus-infected ones; 94 genes were upregulated in the wMelCS112-infected flies and 218 in wMelPlus-infected ones. The most notable genetic difference between the two strains was a large inversion. We find it most likely that such an inversion can significantly modify established genetic communications between the host and symbiont.
Perhaps the most importance aspect of our study were the changes in the expression levels of several stress-associated genes and genes encoding different homologs of alkaline phosphatase (Table 2, Figure 8 and Figure 9). The downregulation of the expression of three genes belonging to the Turandot family and upregulation of two Bomanins connected with them in both infected strains provide evidence for both the suppression of tolerance to long-lasting heat stress and activation of antimicrobial activity. Turandot genes are known to be part of a mechanism determining heat tolerance in Drosophila [42,43], and Bomanin genes have been shown to mediate immunity in Drosophila [44,45]. The latter agrees with data on the increased resistance of Wolbachia-infected insects to bacterial and fungal infections [8]. However, we believe that the most interesting result was the upregulation of the corazonin receptor gene, CrzR, in Bi90wMelPlus flies, characterized by an increased resistance to acute heat stress [11,15]. Corazonin is a neuropeptide related to a vertebrate gonadotropin-releasing hormone and is known to alter stress physiology in Drosophila, mediating metabolic, osmotic and oxidative stress [46,47,48]. CrzR encodes a member of the Class A family of the G-protein-coupled receptor superfamily that transduces the corazonin signal in the salivary glands, the adult fat body and in seven pairs of corazonin neurons in the adult brain [48,49]. It was shown that the knockdown of CrzR in the adult fat body led to a compensatory increase in the corazonin peptide, which, in turn, resulted in an increase in stress resistance in D. melanogaster [48]. It seems that upregulation of CrzR in wMelPlus-infected flies produces a similar effect by increasing sensitivity to the corazonin signal.
Increased expression of the phu and Alp4 genes in Bi90wMelPlus flies and the Alp2, Alp9 and Alp10 genes in Bi90wMelCS112 flies correlated with increased alkaline phosphatase activity demonstrated in them [15] (Figure 12). Notably, the only Alp gene shown to be expressed in the adult brain, Alp4 [50,51], was changed in Bi90wMelPlus flies only, which supports our previous hypothesis [11] that increased stress resistance in wMelPlus-infected flies is connected with changes in dopamine metabolism. Dopamine is known to be produced in the insect brain and to participate in the neuroendocrine stress response [52,53]. It was shown that an elevated level of dopamine correlated with a decrease in resistance to acute heat stress [54]. It was also demonstrated that only one of the known mutations of the Alp4 gene resulted in an abnormal stress response [55]. It seems that Alp4 could encode an enzyme involved in dopamine synthesis, while the Alp2, Alp9 and Alp10 genes encode enzymes not related to dopamine metabolism and stress response. The increase in their activity shown in the flies infected with the Wolbachia strains of the MelCS group, other than wMelPlus [15], does not affect the stress resistance.

4. Materials and Methods

4.1. Drosophila Lines and Rearing

Ten-day-old females from three D. melanogaster lines were used in the study: inbred wild-type line Bi90T treated with tetracycline for three generations to make it Wolbachia-free prior to the start of the experiments [11] and two conplastic lines, carrying the nuclear background of the Bi90T line and cytoplasmic backgrounds with the Wolbachia strains, wMelCS112 or wMelPlus. Lines Bi90CS112 and Bi90wMelPlus were produced by backcrossing Wolbachia donor females with Bi90T males for 20 generations, as described earlier [15]. The corresponding Wolbachia donor lines, 3–112 and w153, were isolated from nature and characterized for any Wolbachia infections [15,32], which was identified as the wMelCS genotype, according to Riegler et al. [9]. However, sequencing and comparative genomic analysis of the wMelPlus and wMelCS112 genomes revealed a large chromosomal inversion in wMelPlus, distinguishing it from wMelCS112 and other known representatives of the wMelCS group of Wolbachia genotypes [17].
Flies were maintained on standard food (agar–agar, 7 g/L; corn grits, 50 g/L; dry yeast, 18 g/L; sugar, 40 g/L) in an MIR-554 incubator (Sanyo, Osaka, Japan) at 25 °C under a 12:12 h light–dark cycle; females were used for the experiments at the age of 10 days.

4.2. RNA Isolation, cDNA Library Construction and RNA Sequencing

Three (for the Bi90wMelCS112 and Bi90wMelPlus lines) and four (for the Bi90T line) independent biological replicates were obtained by RNA extraction from whole females (20 females per sample) collected on the same day for the first (main) batch. For the second (supplementary) batch, four (for the Bi90wMelCS112 line) and five (for the Bi90T and Bi90wMelPlus lines) independent biological replicates were obtained in a similar way. The total RNA was extracted using TRI Reagent (T-9424, Sigma, San Diego, CA, USA) according to the manufacturer’s instructions.
The total RNA quality was assessed using the Agilent Bioanalyzer 2100 system (Agilent Technologies, Santa Clara, CA, USA) with the RNA 6000 Nano Kit (Agilent Technologies, Santa Clara, CA, USA). For the RNA-seq library preparation, 1 μg of high-quality RNA per sample was used as the input material. mRNA fractions were isolated and barcoded. RNA-seq libraries for the Illumina system were constructed using the TruSeq Stranded mRNA Library Prep Kit (Illumina, San Diego, CA, USA) according to the manufacturer’s instructions. The quantity and quality of the libraries were assessed using the Agilent Bioanalyzer 2100 system (Agilent Technologies, Santa Clara, CA, USA) and the DNA High Sensitivity Kit (Agilent Technologies, Santa Clara, CA, USA). The libraries were sequenced using the Illumina NextSeq 550 with 75-bp read length and a sequencing depth of 20 million reads per library.

4.3. Transcriptome Assembly and Quantification

Quality control was performed using FastQC v0.11.5 [56]. Adapters and primers were removed in Trimmomatic v.0.3 [57] using ILLUMINACLIP parameters (with built-in SE-library of TruSeq3-SE index adapters). ILLUMINACLIP:TruSeq3-SE.fa:2:30:10 LEADING:3 TRAILING:3 SLIDINGWINDOW:3:15 MINLEN:75 CROP:75 parameters were used to remove short and low-quality reads. The contig assembly was performed using Hisat2 [58] with the default settings on merged libraries with the annotated genome (GCF_000001215.4). A sorted BAM file was made using SamTools [59]. The quantification was performed using the featureCounts program [27]. The supplementary batch was mapped and quantified using the kallisto algorithm [60].

4.4. Gene Annotation and Analysis of Differential Expression

Annotation of genes and biochemical pathways was done using ShinyGOv77 [61] and the Flybase database [62]. Differential expression analysis was performed with the use of the edgeR v. 4.3 software package [63]. Results with an FDR parameter value of less than 0.05 were selected as differentially expressed genes. All DEGs, regardless of the log2 fold change value, remained included to maximize information retention. Log2 fold changes were used in all analyses and figures, except heat maps; counts per million (CPM) were used there instead.

4.5. Quantitative Real-Time Polymerase Chain Reaction

For quantitative real-time polymerase chain reaction (qRT-PCR) analysis, the RNA extraction procedure was identical to that for RNA sequencing. The synthesis of cDNA was carried out using the ABScript III RT Master Mix for qPCR with gDNA Remover #RK20429 (Abclonal Technology, Woburn, MA, USA). The expression of genes under study was analyzed on the CFX96 (Bio-Rad, Hercules, CO, USA) with qRT-PCR using three replicates for every sample. The qPCR mix contained R-402 c SYBR-Green I (Syntol, Moscow, Russia). Each reaction was performed with three biological replicates. The expression was quantified using the relative 2−∆∆Ct method [64,65] with internal control primers specific for β-Tubulin and Actin 5C [65]. Primer sequences are given in Table 5.

4.6. Statistical Analysis

Principal components were calculated from a set of expression level values of DEGs (714 genes) for ten samples. The expression profiles were filtered to exclude null values. Next, for each gene, the expression profile was centered and normalized. Any excess of the number of traits over the number of objects was not an obstacle; the calculation of principal components can be done through an SVD transformation of the object–trait matrix at any ratios for the number of objects and traits. The analysis was performed by using the Past package v4.13 [66].
When conducting the qRT-PCR, three biological replicates were used, each of which was obtained from three technical ones. The CFX96 System (Bio-Rad, USA) reports only the mean of three technical values and the mean error, so it is impossible to check normality, use nonparametric tests or even apply bootstrap. However, from this information, it is easy to obtain the sum of the squares of the three technical values. This was enough to calculate both the overall mean and its error. The number of technical values for each overall mean was 9. When calculating the Student’s t-test, we obtained 2 × 9 − 2 = 16 degrees of freedom. It is known that the criteria of significance, such as in the Student’s t-test, are resistant to deviations from normality [67] due to the distribution of means approaching normality with an increasing sample size. Since a total of 10 comparisons were made, the Benjamini–Hochberg correction was additionally calculated for the p-value to compare with three standard significance levels [68].

5. Conclusions

In this study, we aimed to investigate the following aspects of host–bacteria interaction: the effects of the presence of the cytoplasmically-transmitted bacteria Wolbachia on the insect host’s gene expression patterns and a possible correlation of these effects with previously described changes in host fitness and metabolism caused by Wolbachia. We employed a transcriptomics approach to investigate how Wolbachia affects differential expression of genes in D. melanogaster females and whether a large chromosomal inversion in the bacterial genome influences the manner in which the host’s gene expression is altered by the Wolbachia infection. Our analyses revealed 493 DEGs that exhibit significant changes in expression in the flies infected with the wMelCS112 Wolbachia strain, which is a typical representative of the wMelCS group of genotypes, and 336 DEGs that exhibit significant changes in expression in the flies infected with the wMelPlus Wolbachia strain, which differed from other representatives of the wMelCS group by an inversion. A total of 368 DEGs and 236 DEGs from these two groups, respectively, were linked into gene networks, in which the following functional groups were designated based on DEGs annotations: “Proteolysis”, “Carbohydrate transport and metabolism”, “Oxidation–reduction process”, “Embryogenesis”, “Transmembrane transport”, “Response to stress” and “Alkaline phosphatases”. Thus, we demonstrated that the Wolbachia infection contributed to differential expression of host genes in a significant way. We also found that the list of differentially expressed genes differed between the wMelPlus-infected and wMelCS112-infected females, which indicates the important role of the bacterial genotype in Wolbachia–host interactions. This suggestion is also supported by the fact that the wMelPlus infection results in more than twice the number of upregulated DEGs and half the number of downregulated DEGs compared to the wMelCS112 infection. Notably, the only stress-related gene, the expression of which was found to be increased in stress-resistant flies (infected with wMelPlus) compared to control flies (uninfected and infected with the wMelSC112 strain), was the corazonin receptor gene, CrzR. In the future, we plan to research the role of the CrzR gene in providing resistance to factors such as heat stress in Wolbachia-infected flies.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms242417411/s1.

Author Contributions

Conceptualization, N.E.G.; methodology, O.V.A., M.A.B., M.A.D. and G.V.V.; software, M.A.D.; validation, O.V.A. and G.V.V.; formal analysis, M.A.D., N.E.G. and O.D.S.; investigation, O.V.A., M.A.B., G.V.V. and N.V.S.; data curation, M.A.D.; writing—original draft preparation, O.V.A., O.D.S., N.E.G., G.V.V. and M.A.D.; writing—review and editing, N.E.G. and O.D.S.; visualization, N.E.G. and M.A.D.; supervision, N.E.G.; project administration, N.E.G.; funding acquisition, N.E.G. All authors have read and agreed to the published version of the manuscript.

Funding

The study is supported by RSF grant No. 21-14-00090.

Institutional Review Board Statement

The study was conducted according to the protocols approved by the Committee on the Ethics of Animal Experiments of the ICG SB RAS (Permit Number: 7 of 19 November 2011).

Informed Consent Statement

Not applicable.

Data Availability Statement

Data have been deposited in the GEO database; the accession numbers have not yet been obtained and will be provided during the review.

Acknowledgments

We thank Vadim M. Efimov for advice on the statistical analysis and Darya Kochetova for language editing. Experimental D. melanogaster lines in the Drosophila collection of the Institute of Cytology and Genetics SB RAS were maintained with the help of the Ministry of Science and Higher Education of the Russian Federation (#FWNR-2022-0019).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Hilgenboecker, K.; Hammerstein, P.; Schlattmann, P.; Telschow, A.; Werren, J.H. How many species are infected with Wolbachia?—A statistical analysis of current data. FEMS Microbiol. Lett. 2008, 281, 215–220. [Google Scholar] [CrossRef] [PubMed]
  2. Zug, R.; Hammerstein, P. Still a host of hosts for Wolbachia: Analysis of recent data suggests that 40% of terrestrial arthropod species are infected. PLoS ONE 2012, 7, e38544. [Google Scholar] [CrossRef] [PubMed]
  3. Min, K.T.; Benzer, S. Wolbachia, normally a symbiont of Drosophila, can be virulent, causing degeneration and early death. Proc. Natl. Acad. Sci. USA 1997, 94, 10792–10796. [Google Scholar] [CrossRef]
  4. Graham, R.I.; Grzywacz, D.; Mushobozi, W.L.; Wilson, K. Wolbachia in a major African crop pest increases susceptibility toviral disease rather than protects. Ecol. Lett. 2012, 15, 993–1000. [Google Scholar] [CrossRef] [PubMed]
  5. Strunov, A.A.; Ilinskii, Y.Y.; Zakharov, I.K.; Kiseleva, E.V. Effect of high temperature on survival of Drosophila melanogaster infected with pathogenic strain of Wolbachia bacteria. Russ. J. Genet. Appl. Res. 2013, 3, 435–443. [Google Scholar] [CrossRef]
  6. Martinez, J.; Ok, S.; Smith, S.; Snoeck, K.; Day, J.P.; Jiggins, F.M. Should symbionts be nice or selfish? Antiviral effects of Wolbachia are costly but reproductive parasitism is not. PLoS Pathog. 2015, 11, e1005021. [Google Scholar] [CrossRef]
  7. Werren, J.H.; Baldo, L.; Clark, M.E. Wolbachia: Master manipulators of invertebrate biology. Nat. Rev. Microbiol. 2008, 6, 741–751. [Google Scholar] [CrossRef]
  8. Burdina, E.V.; Gruntenko, N.E. Physiological Aspects of Wolbachia pipientis–Drosophila melanogaster Relationship. J. Evol. Biochem. Phys. 2022, 58, 303–317. [Google Scholar] [CrossRef]
  9. Riegler, M.; Sidhu, M.; Miller, W.J.; O’Neill, S.L. Evidence for a global Wolbachia replacement in Drosophila melanogaster. Curr. Biol. 2005, 15, 1428–1433. [Google Scholar] [CrossRef]
  10. Martinez, J.; Tolosana, I.; Ok, S.; Smith, S.; Snoeck, K.; Day, J.P.; Jiggins, F.M. Symbiont strain is the main determinant of variation in Wolbachia-mediated protection against viruses across Drosophila species. Mol. Ecol. 2017, 26, 4072–4084. [Google Scholar] [CrossRef]
  11. Gruntenko, N.E.; Ilinsky, Y.Y.; Adonyeva, N.V.; Burdina, E.V.; Bykov, R.A.; Menshanov, P.N.; Rauschenbach, I.Y. Various Wolbachia genotypes differently influence host Drosophila dopamine metabolism and survival under heat stress conditions. BMC Evol. Biol. 2017, 17 (Suppl. S2), 252. [Google Scholar] [CrossRef] [PubMed]
  12. Gruntenko, N.E.; Karpova, E.K.; Adonyeva, N.V.; Andreenkova, O.V.; Burdina, E.V.; Ilinsky, Y.Y.; Bykov, R.A.; Menshanov, P.N.; Rauschenbach, I.Y. Drosophila female fertility and juvenile hormone metabolism depends on the type of Wolbachia infection. J. Exp. Biol. 2019, 222, jeb195347. [Google Scholar] [CrossRef]
  13. Fry, A.J.; Rand, D.M. Wolbachia interactions that determine Drosophila melanogaster survival. Evolution 2002, 56, 1976–1981. [Google Scholar]
  14. Dean, M.D. A Wolbachia-associated fitness benefit depends on genetic background in Drosophila simulans. Proc. R. Soc. B Biol. Sci. 2006, 273, 1415–1420. [Google Scholar] [CrossRef] [PubMed]
  15. Burdina, E.V.; Bykov, R.A.; Menshanov, P.N.; Ilinsky, Y.Y.; Gruntenko, N.E. Unique Wolbachia strain wMelPlus increases heat stress resistance in Drosophila melanogaster. Arch. Insect Biochem. Physiol. 2021, 106, e21776. [Google Scholar] [CrossRef] [PubMed]
  16. Karpova, E.K.; Bobrovskikh, M.A.; Deryuzhenko, M.A.; Shishkina, O.D.; Gruntenko, N.E. Wolbachia Effect on Drosophila melanogaster Lipid and Carbohydrate Metabolism. Insects 2023, 14, 357. [Google Scholar] [CrossRef]
  17. Korenskaia, A.E.; Shishkina, O.D.; Klimenko, A.I.; Andreenkova, O.V.; Bobrovskikh, M.A.; Shatskaya, N.V.; Vasiliev, G.V.; Gruntenko, N.E. New Wolbachia pipientis Genotype Increasing Heat Stress Resistance of Drosophila melanogaster Host Is Characterized by a Large Chromosomal Inversion. Int. J. Mol. Sci. 2022, 23, 16212. [Google Scholar] [CrossRef]
  18. Cui, L.; Neoh, H.M.; Iwamoto, A.; Hiramatsu, K. Coordinated phenotype switching with large-scale chromosome flip-flop inversion observed in bacteria. Proc. Natl. Acad. Sci. USA 2012, 109, e1647-56. [Google Scholar] [CrossRef]
  19. Chrostek, E.; Teixeira, L. Mutualism Breakdown by Amplification of Wolbachia Genes. PLoS Biol. 2015, 13, e1002065. [Google Scholar] [CrossRef]
  20. Duarte, E.H.; Carvalho, A.; López-Madrigal, S.; Costa, J.; Teixeira, L. Forward genetics in Wolbachia: Regulation of Wolbachia proliferation by the amplification and deletion of an addictive genomic island. PLoS Genet. 2021, 17, e1009612. [Google Scholar] [CrossRef]
  21. Lindsey, A.R.I. Sensing, signaling, and secretion: A review and analysis of systems for regulating host interaction in Wolbachia. Genes 2020, 11, 813. [Google Scholar] [CrossRef] [PubMed]
  22. Mayoral, J.G.; Hussain, M.; Joubert, D.A.; Iturbe-Ormaetxe, I.; O’Neill, S.L.; Asgari, S. Wolbachia small noncoding RNAs and their role in cross-kingdom communications. Proc. Natl. Acad. Sci. USA 2014, 111, 18721–18726. [Google Scholar] [CrossRef] [PubMed]
  23. Ferree, P.M.; Frydman, H.M.; Li, J.M.; Cao, J.; Wieschaus, E.; Sullivan, W. Wolbachia Utilizes Host Microtubules and Dynein for Anterior Localization in the Drosophila Oocyte. PLoS Pathog. 2005, 1, e14. [Google Scholar] [CrossRef]
  24. Mateos, M.; Silva, N.O.; Ramirez, P.; Higareda-Alvear, V.M.; Aramayo, R.; Erickson, J.W. Effect of heritable symbionts on maternally-derived embryo transcripts. Sci. Rep. 2019, 9, 8847. [Google Scholar] [CrossRef] [PubMed]
  25. Frantz, S.I.; Small, C.M.; Cresko, W.A.; Singh, N.D. Ovarian transcriptional response to Wolbachia infection in D. melanogaster in the context of between-genotype variation in gene expression. G3 Genes Genomes Genet. 2023, 3, jkad047. [Google Scholar] [CrossRef] [PubMed]
  26. Goh, W.W.B.; Wang, W.; Wong, L. Why batch effects matter in omics data, and how to avoid them. Trends Biotechnol. 2017, 35, 498–507. [Google Scholar] [CrossRef]
  27. Liao, Y.; Smyth, G.K.; Shi, W. featureCounts: An efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics 2014, 30, 923–930. [Google Scholar] [CrossRef]
  28. Lindsey, A.R.I.; Bhattacharya, T.; Hardy, R.W.; Newton, I.L.G. Wolbachia and Virus Alter the Host Transcriptome at the Interface of Nucleotide Metabolism Pathways. mBio 2021, 12, e03472-20. [Google Scholar] [CrossRef]
  29. Detcharoen, M.; Schilling, M.P.; Arthofer, W.; Schlick-Steiner, B.C.; Steiner, F.M. Differential gene expression in Drosophila melanogaster and D. nigrosparsa infected with the same Wolbachia strain. Sci. Rep. 2021, 11, 11336. [Google Scholar] [CrossRef] [PubMed]
  30. He, Z.; Zheng, Y.; Yu, W.J.; Fang, Y.; Mao, B.; Wang, Y.F. How do Wolbachia modify the Drosophila ovary? New evidences support the "titration-restitution" model for the mechanisms of Wolbachia-induced CI. BMC Genom. 2019, 20, 608. [Google Scholar] [CrossRef]
  31. Nunes, M.D.; Nolte, V.; Schlotterer, C. Nonrandom Wolbachia infection status of Drosophila melanogaster strains with different mtDNA haplotypes. Mol. Biol. Evol. 2008, 25, 2493–2498. [Google Scholar] [CrossRef] [PubMed]
  32. Ilinsky, Y.Y. Coevolution of Drosophila melanogaster mtDNA and Wolbachia genotypes. PLoS ONE 2013, 8, e54373. [Google Scholar] [CrossRef]
  33. Bykov, R.A.; Yudina, M.A.; Gruntenko, N.E.; Zakharov, I.K.; Voloshina, M.A.; Melashchenko, E.S.; Danilova, M.V.; Mazunin, I.O.; Ilinsky, Y.Y. Prevalence and genetic diversity of Wolbachia endosymbiont and mtDNA in Palearctic populations of Drosophila melanogaster. BMC Evol. Biol. 2019, 19 (Suppl. S1), 48. [Google Scholar] [CrossRef] [PubMed]
  34. Reynolds, K.T.; Thomson, L.J.; Hoffmann, A.A. The effects of host age, hostnuclear background and temperature on phenotypic effects of the virulentWolbachia strain popcorn in Drosophila melanogaster. Genetics 2003, 164, 1027–1034. [Google Scholar] [CrossRef] [PubMed]
  35. Woolfit, M.; Iturbe-Ormaetxe, I.; Brownlie, J.C.; Walker, T.; Riegler, M.; Seleznev, A.; Popovici, J.; Rancès, E.; Wee, B.A.; Pavlides, J.; et al. Genomic evolution of the pathogenic Wolbachia strain, wMelPop. Genome Biol. Evol. 2013, 5, 2189–2204. [Google Scholar] [CrossRef]
  36. Dou, W.; Miao, Y.; Xiao, J.; Huang, D. Association of Wolbachia with Gene Expression in Drosophila Testes. Microb. Ecol. 2021, 82, 805–817. [Google Scholar] [CrossRef]
  37. Zhang, H.B.; Cao, Z.; Qiao, J.X.; Zhong, Z.Q.; Pan, C.C.; Liu, C.; Zhang, L.M.; Wang, Y.F. Metabolomics provide new insights into mechanisms of Wolbachia-induced paternal defects in Drosophila melanogaster. PLoS Pathog. 2021, 17, e1009859. [Google Scholar] [CrossRef]
  38. Roy, S.; Saha, T.T.; Zou, Z.; Raikhel, A.S. Regulatory Pathways Controlling Female Insect Reproduction. Annu. Rev. Entomol. 2018, 63, 489–511. [Google Scholar] [CrossRef]
  39. Melnikow, E.; Xu, S.; Liu, J.; Bell, A.J.; Ghedin, E.; Unnasch, T.R.; Lustigman, S. A potential role for the interaction of Wolbachia surface proteins with the Brugia malayi glycolytic enzymes and cytoskeleton in maintenance of endosymbiosis. PLoS Negl. Trop. Dis. 2013, 7, e2151. [Google Scholar] [CrossRef]
  40. Ponton, F.; Wilson, K.; Holmes, A.; Raubenheimer, D.; Robinson, K.L.; Simpson, S.J. Macronutrients mediate the functional relationship between Drosophila and Wolbachia. Proc. Biol. Sci. 2015, 282, 20142029. [Google Scholar] [CrossRef]
  41. Kryukova, N.A.; Kryukov, V.Y.; Polenogova, O.V.; Chertkova, E.A.; Tyurin, M.V.; Rotskaya, U.N.; Alikina, T.; Kabilov, M.R.; Glupov, V.V. The endosymbiotic bacterium Wolbachia (Rickettsiales) alters larval metabolism of the parasitoid Habrobracon hebetor (Hymenoptera: Braconidae). Arch. Insect Biochem. Physiol. 2023, 114, e22053. [Google Scholar] [CrossRef] [PubMed]
  42. Gruntenko, N.E.; Karpova, E.K.; Babenko, V.N.; Vasiliev, G.V.; Andreenkova, O.V.; Bobrovskikh, M.A.; Menshanov, P.N.; Babenko, R.O.; Rauschenbach, I.Y. Fitness Analysis and Transcriptome Profiling Following Repeated Mild Heat Stress of Varying Frequency in Drosophila melanogaster Females. Biology 2021, 10, 1323. [Google Scholar] [CrossRef] [PubMed]
  43. Amstrup, A.B.; Bæk, I.; Loeschcke, V.; Givskov Sørensen, J. A functional study of the role of Turandot genes in Drosophila melanogaster: An emerging candidate mechanism for inducible heat tolerance. J. Insect Physiol. 2022, 143, 104456. [Google Scholar] [CrossRef] [PubMed]
  44. Clemmons, A.W.; Lindsay, S.A.; Wasserman, S.A. An effector Peptide family required for Drosophila toll-mediated immunity. PLoS Pathog. 2015, 11, e1004876. [Google Scholar] [CrossRef]
  45. Lindsay, S.A.; Lin, S.J.H.; Wasserman, S.A. Short-Form Bomanins Mediate Humoral Immunity in Drosophila. J. Innate Immun. 2018, 10, 306–314. [Google Scholar] [CrossRef] [PubMed]
  46. Veenstra, J.A. Does corazonin signal nutritional stress in insects? Insect Biochem. Mol. Biol. 2009, 39, 755–762. [Google Scholar] [CrossRef] [PubMed]
  47. Zandawala, M.; Nguyen, T.; Balanyà Segura, M.; Johard, H.A.D.; Amcoff, M.; Wegener, C.; Paluzzi, J.P.; Nässel, D.R. A neuroendocrine pathway modulating osmotic stress in Drosophila. PLoS Genet. 2021, 17, e1009425. [Google Scholar] [CrossRef]
  48. Kubrak, O.I.; Lushchak, O.V.; Zandawala, M.; Nässel, D.R. Systemic corazonin signalling modulates stress responses and metabolism in Drosophila. Open Biol. 2016, 6, 160152. [Google Scholar] [CrossRef]
  49. Lin, H.H.; Kuang, M.C.; Hossain, I.; Xuan, Y.; Beebe, L.; Shepherd, A.K.; Rolandi, M.; Wang, J.W. A nutrient-specific gut hormone arbitrates between courtship and feeding. Nature 2022, 602, 632–638. [Google Scholar] [CrossRef]
  50. Yang, M.Y.; Wang, Z.; MacPherson, M.; Dow, J.A.; Kaiser, K. A novel Drosophila alkaline phosphatase specific to the ellipsoid body of the adult brain and the lower Malpighian (renal) tubule. Genetics 2000, 154, 285–297. [Google Scholar] [CrossRef]
  51. Aradska, J.; Bulat, T.; Sialana, F.J.; Birner-Gruenberger, R.; Erich, B.; Lubec, G. Gel-free mass spectrometry analysis of Drosophila melanogaster heads. Proteomics 2015, 15, 3356–3360. [Google Scholar] [CrossRef] [PubMed]
  52. Gruntenko, N.E.; Rauschenbach, I.Y. Interplay of JH, 20E and biogenic amines under normal and stress conditions and its effects on reproduction. J. Insect Physiol. 2008, 54, 902–908. [Google Scholar] [CrossRef] [PubMed]
  53. Hanna, M.E.; Bednářová, A.; Rakshit, K.; Chaudhuri, A.; O’Donnell, J.M.; Krishnan, N. Perturbations in dopamine synthesis lead to discrete physiological effects and impact oxidative stress response in drosophila. J. Insect Physiol. 2015, 73, 11–19. [Google Scholar] [CrossRef] [PubMed]
  54. Gruntenko, N.E.; Karpova, E.K.; Alekseev, A.A.; Chentsova, N.A.; Saprykina, Z.V.; Bownes, M.; Rauschenbach, I.Y. Effects of dopamine on juvenile hormone metabolism and fitness in Drosophila virilis. J. Insect Physiol. 2005, 51, 959–968. [Google Scholar] [CrossRef] [PubMed]
  55. Khan, S.J.; Abidi, S.N.F.; Skinner, A.; Tian, Y.; Smith-Bolton, R.K. The Drosophila Duox maturation factor is a key component of a positive feedback loop that sustains regeneration signaling. PLoS Genet. 2017, 13, e1006937. [Google Scholar] [CrossRef] [PubMed]
  56. Babraham Bioinformatics FastQC. Available online: https://www.bioinformatics.babraham.ac.uk/projects/fastqc/ (accessed on 30 October 2023).
  57. Anthony, M.B.; Lohse, M.; Usadel, B. Trimmomatic: A flexible trimmer for Illumina sequence data. Bioinformatics 2014, 30, 2114–2120. [Google Scholar]
  58. Kim, D.; Langmead, B.; Salzberg, S. HISAT: A fast spliced aligner with low memory requirements. Nat. Methods 2015, 12, 357–360. [Google Scholar] [CrossRef]
  59. Li, H.; Handsaker, B.; Wysoker, A.; Fennell, T.; Ruan, J.; Homer, N.; Marth, G.; Abecasis, G.; Durbin, R. The Sequence Alignment/Map format and SAMtools. Bioinformatics 2009, 25, 2078–2079. [Google Scholar] [CrossRef]
  60. Bray, N.L.; Pimentel, H.; Melsted, P.; Pachter, L. Near-optimal probabilistic RNA-seq quantification. Nat. Biotechnol. 2016, 34, 525–527. [Google Scholar] [CrossRef]
  61. Ge, S.H.; Jung, D.; Yao, R. ShinyGO: A graphical gene-set enrichment tool for animals and plants. Bioinformatics 2020, 36, 2628–2629. [Google Scholar] [CrossRef]
  62. Thurmond, J.; Goodman, J.L.; Strelets, V.B.; Attrill, H.; Gramates, L.S.; Marygold, S.J.; Matthews, B.B.; Millburn, G.; Antonazzo, G.; Trovisco, V.; et al. FlyBase 2.0: The next generation. Nucleic Acids Res. 2019, 47, D759–D765. [Google Scholar] [CrossRef]
  63. Robinson, M.D.; McCarthy, D.J.; Smyth, G.K. edgeR: A Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 2010, 26, 139–140. [Google Scholar] [CrossRef] [PubMed]
  64. Schmittgen, T.D.; Livak, K.J. Analyzing real-time PCR data by the comparative C(T) method. Nat. Protoc. 2008, 3, 1101–1108. [Google Scholar] [CrossRef] [PubMed]
  65. Ponton, F.; Chapuis, M.P.; Pernice, M.; Sword, G.A.; Simpson, S.J. Evaluation of potential reference genes for reverse transcription-qPCR studies of physiological responses in Drosophila melanogaster. J. Insect Physiol. 2011, 57, 840–850. [Google Scholar] [CrossRef] [PubMed]
  66. Hammer, Ø.; Harper, D.A.T. Past: Paleontological statistics software package for educaton and data anlysis. Palaeontol. Electron. 2001, 4, 1–9. [Google Scholar]
  67. Kendall, M.G.; Stuart, A. The Advanced Theory of Statistics: Inference and Relationship, 2nd ed.; Hafner Publishing Company: New York, NY, USA, 1961; Volume 2, 312p. [Google Scholar]
  68. Narkevich, A.N.; Vinogradov, K.A.; Grjibovski, A.M. Multiple Comparisons in Biomedical Research: The Problem and its Solutions. Ekol. Cheloveka 2020, 10, 55–64. [Google Scholar] [CrossRef]
Figure 1. Principal component analysis of the RNA-seq data distribution. Components were calculated from a set of expression level values of DEGs (714 genes) for ten samples. The first and second components describe 77.3% of the variants. The uninfected Bi90T line differs from the infected lines by PC2; the wMelPlus-infected line differs from the two other lines under study by PC1.
Figure 1. Principal component analysis of the RNA-seq data distribution. Components were calculated from a set of expression level values of DEGs (714 genes) for ten samples. The first and second components describe 77.3% of the variants. The uninfected Bi90T line differs from the infected lines by PC2; the wMelPlus-infected line differs from the two other lines under study by PC1.
Ijms 24 17411 g001
Figure 2. Gene networks for differentially expressed genes in wMelPlus-infected females compared to uninfected ones. The functional groups are enclosed in circles of different colors. The color of the node shows the direction and level of gene expression change in wMelPlus-infected females compared to uninfected ones. Color gradient from blue to red shows the expression level of genes relative to each other. Shades of blue indicate a decrease in expression; shades of red indicate an increase in expression. STRING was used to identify the core network of interactions at a confidence threshold of 0.4 (medium confidence).
Figure 2. Gene networks for differentially expressed genes in wMelPlus-infected females compared to uninfected ones. The functional groups are enclosed in circles of different colors. The color of the node shows the direction and level of gene expression change in wMelPlus-infected females compared to uninfected ones. Color gradient from blue to red shows the expression level of genes relative to each other. Shades of blue indicate a decrease in expression; shades of red indicate an increase in expression. STRING was used to identify the core network of interactions at a confidence threshold of 0.4 (medium confidence).
Ijms 24 17411 g002
Figure 3. Gene networks for differentially expressed genes in wMelCS112-infected females compared to uninfected ones. The functional groups are enclosed in circles of different colors. The color of the node shows the direction and level of gene expression change in wMelCS112-infected females compared to uninfected ones. Color gradient from blue to red shows the expression level of genes relative to each other. Shades of blue indicate a decrease in expression; shades of red indicate an increase in expression. STRING was used to identify the core network of interactions at a confidence threshold of 0.4 (medium confidence).
Figure 3. Gene networks for differentially expressed genes in wMelCS112-infected females compared to uninfected ones. The functional groups are enclosed in circles of different colors. The color of the node shows the direction and level of gene expression change in wMelCS112-infected females compared to uninfected ones. Color gradient from blue to red shows the expression level of genes relative to each other. Shades of blue indicate a decrease in expression; shades of red indicate an increase in expression. STRING was used to identify the core network of interactions at a confidence threshold of 0.4 (medium confidence).
Ijms 24 17411 g003
Figure 4. Heat map of differentially expressed genes belonging to the “Carbohydrate transport and metabolism” functional group in flies infected with wMelPlus and wMelCS112 Wolbachia strains compared to uninfected flies. Uninfected_1, Uninfected_2, Uninfected_3—gene expression of the uninfected Drosophila melanogaster line; wMelCS112_1, wMelCS112_2, wMelCS112_3—the Drosophila melanogaster line infected with wMelCS112; wMelPlus_1, wMelPlus_2, wMelPlus_3—the Drosophila melanogaster line infected with wMelPlus. Genes were clustered using the linkage method. The heat maps were generated using the Python3 package seaborn v0.13.0.
Figure 4. Heat map of differentially expressed genes belonging to the “Carbohydrate transport and metabolism” functional group in flies infected with wMelPlus and wMelCS112 Wolbachia strains compared to uninfected flies. Uninfected_1, Uninfected_2, Uninfected_3—gene expression of the uninfected Drosophila melanogaster line; wMelCS112_1, wMelCS112_2, wMelCS112_3—the Drosophila melanogaster line infected with wMelCS112; wMelPlus_1, wMelPlus_2, wMelPlus_3—the Drosophila melanogaster line infected with wMelPlus. Genes were clustered using the linkage method. The heat maps were generated using the Python3 package seaborn v0.13.0.
Ijms 24 17411 g004
Figure 5. Heat map of differentially expressed genes belonging to the “Oxidation–reduction process” functional group in flies infected with wMelPlus and wMelCS112 Wolbachia strains compared to uninfected flies. Uninfected_1, Uninfected_2, Uninfected_3—gene expression of the uninfected Drosophila melanogaster line; wMelCS112_1, wMelCS112_2, wMelCS112_3—the Drosophila melanogaster line infected with wMelCS112; wMelPlus_1, wMelPlus_2, wMelPlus_3—the Drosophila melanogaster line infected with wMelPlus. Genes were clustered using the linkage method. The heat maps were generated using the Python3 package seaborn v0.13.0.
Figure 5. Heat map of differentially expressed genes belonging to the “Oxidation–reduction process” functional group in flies infected with wMelPlus and wMelCS112 Wolbachia strains compared to uninfected flies. Uninfected_1, Uninfected_2, Uninfected_3—gene expression of the uninfected Drosophila melanogaster line; wMelCS112_1, wMelCS112_2, wMelCS112_3—the Drosophila melanogaster line infected with wMelCS112; wMelPlus_1, wMelPlus_2, wMelPlus_3—the Drosophila melanogaster line infected with wMelPlus. Genes were clustered using the linkage method. The heat maps were generated using the Python3 package seaborn v0.13.0.
Ijms 24 17411 g005
Figure 6. Heat map of differentially expressed genes belonging to the “Embryogenesis” functional group in flies infected with wMelPlus and wMelCS112 Wolbachia strains compared to uninfected flies. Uninfected_1, Uninfected_2, Uninfected_3—gene expression of the uninfected Drosophila melanogaster line; wMelCS112_1, wMelCS112_2, wMelCS112_3—the Drosophila melanogaster line infected with wMelCS112; wMelPlus_1, wMelPlus_2, wMelPlus_3—the Drosophila melanogaster line infected with wMelPlus. Genes were clustered using the linkage method. The heat maps were generated using the Python3 package seaborn v0.13.0.
Figure 6. Heat map of differentially expressed genes belonging to the “Embryogenesis” functional group in flies infected with wMelPlus and wMelCS112 Wolbachia strains compared to uninfected flies. Uninfected_1, Uninfected_2, Uninfected_3—gene expression of the uninfected Drosophila melanogaster line; wMelCS112_1, wMelCS112_2, wMelCS112_3—the Drosophila melanogaster line infected with wMelCS112; wMelPlus_1, wMelPlus_2, wMelPlus_3—the Drosophila melanogaster line infected with wMelPlus. Genes were clustered using the linkage method. The heat maps were generated using the Python3 package seaborn v0.13.0.
Ijms 24 17411 g006
Figure 7. Heat map of differentially expressed genes belonging to the “Transmembrane transport” functional group in flies infected with wMelPlus and wMelCS112 Wolbachia strains compared to uninfected flies. Uninfected_1, Uninfected_2, Uninfected_3—gene expression of the uninfected Drosophila melanogaster line; wMelCS112_1, wMelCS112_2, wMelCS112_3—the Drosophila melanogaster line infected with wMelCS112; wMelPlus_1, wMelPlus_2, wMelPlus_3—the Drosophila melanogaster line infected with wMelPlus. Genes were clustered using the linkage method. The heat maps were generated using the Python3 package seaborn v0.13.0.
Figure 7. Heat map of differentially expressed genes belonging to the “Transmembrane transport” functional group in flies infected with wMelPlus and wMelCS112 Wolbachia strains compared to uninfected flies. Uninfected_1, Uninfected_2, Uninfected_3—gene expression of the uninfected Drosophila melanogaster line; wMelCS112_1, wMelCS112_2, wMelCS112_3—the Drosophila melanogaster line infected with wMelCS112; wMelPlus_1, wMelPlus_2, wMelPlus_3—the Drosophila melanogaster line infected with wMelPlus. Genes were clustered using the linkage method. The heat maps were generated using the Python3 package seaborn v0.13.0.
Ijms 24 17411 g007
Figure 8. Heat map of differentially expressed genes belonging to the “Response to stress” functional group in flies infected with wMelPlus and wMelCS112 Wolbachia strains compared to uninfected flies. Uninfected_1, Uninfected_2, Uninfected_3—gene expression of the uninfected Drosophila melanogaster line; wMelCS112_1, wMelCS112_2, wMelCS112_3—the Drosophila melanogaster line infected with wMelCS112; wMelPlus_1, wMelPlus_2, wMelPlus_3—the Drosophila melanogaster line infected with wMelPlus. Genes were clustered using the linkage method. The heat maps were generated using the Python3 package seaborn v0.13.0.
Figure 8. Heat map of differentially expressed genes belonging to the “Response to stress” functional group in flies infected with wMelPlus and wMelCS112 Wolbachia strains compared to uninfected flies. Uninfected_1, Uninfected_2, Uninfected_3—gene expression of the uninfected Drosophila melanogaster line; wMelCS112_1, wMelCS112_2, wMelCS112_3—the Drosophila melanogaster line infected with wMelCS112; wMelPlus_1, wMelPlus_2, wMelPlus_3—the Drosophila melanogaster line infected with wMelPlus. Genes were clustered using the linkage method. The heat maps were generated using the Python3 package seaborn v0.13.0.
Ijms 24 17411 g008
Figure 9. Heat map of differentially expressed genes belonging to the “Alkaline phosphatase” functional group in flies infected with wMelPlus and wMelCS112 Wolbachia strains compared to uninfected flies. Uninfected_1, Uninfected_2, Uninfected_3—gene expression of the uninfected Drosophila melanogaster line; wMelCS112_1, wMelCS112_2, wMelCS112_3—the Drosophila melanogaster line infected with wMelCS112; wMelPlus_1, wMelPlus_2, wMelPlus_3—the Drosophila melanogaster line infected with wMelPlus. Genes were clustered using the linkage method. The heat maps were generated using the Python3 package seaborn v0.13.0.
Figure 9. Heat map of differentially expressed genes belonging to the “Alkaline phosphatase” functional group in flies infected with wMelPlus and wMelCS112 Wolbachia strains compared to uninfected flies. Uninfected_1, Uninfected_2, Uninfected_3—gene expression of the uninfected Drosophila melanogaster line; wMelCS112_1, wMelCS112_2, wMelCS112_3—the Drosophila melanogaster line infected with wMelCS112; wMelPlus_1, wMelPlus_2, wMelPlus_3—the Drosophila melanogaster line infected with wMelPlus. Genes were clustered using the linkage method. The heat maps were generated using the Python3 package seaborn v0.13.0.
Ijms 24 17411 g009
Figure 10. Euler diagram of four pairwise comparisons for DEGs included in the functional groups: upregulated DEGs in wMelPlus-infected flies vs. downregulated DEGs in wMelPlus-infected flies, upregulated DEGs in wMelCS112-infected flies vs. downregulated DEGs in wMelCS112-infected flies, upregulated DEGs in wMelPlus-infected flies vs. upregulated DEGs in wMelCS112-infected flies, downregulated DEGs in wMelPlus-infected flies vs. downregulated DEGs in wMelCS112-infected flies. The elliptical semi-transparent shapes indicate sets of DEGs: red ellipse for up-regulated DEGs in wMelPlus-infected flies, blue for down-regulated DEGs in wMelPlus-infected flies, green for up-regulated DEGs in wMelCS112-infected flies, yellow for down-regulated DEGs in wMelCS112-infected flies. The numbers indicate the quantity of genes belonging to one or more intersecting sets of DEGs.
Figure 10. Euler diagram of four pairwise comparisons for DEGs included in the functional groups: upregulated DEGs in wMelPlus-infected flies vs. downregulated DEGs in wMelPlus-infected flies, upregulated DEGs in wMelCS112-infected flies vs. downregulated DEGs in wMelCS112-infected flies, upregulated DEGs in wMelPlus-infected flies vs. upregulated DEGs in wMelCS112-infected flies, downregulated DEGs in wMelPlus-infected flies vs. downregulated DEGs in wMelCS112-infected flies. The elliptical semi-transparent shapes indicate sets of DEGs: red ellipse for up-regulated DEGs in wMelPlus-infected flies, blue for down-regulated DEGs in wMelPlus-infected flies, green for up-regulated DEGs in wMelCS112-infected flies, yellow for down-regulated DEGs in wMelCS112-infected flies. The numbers indicate the quantity of genes belonging to one or more intersecting sets of DEGs.
Ijms 24 17411 g010
Figure 11. Relative gene expression in flies infected with wMelCS112 and wMelPlus Wolbachia strains (lines Bi90wMelCS112 and Bi90wMelPlus, respectively) and uninfected flies of line Bi90T ± SEM. Asterisk indicates differences from the uninfected control. Three asterisks indicate p < 0.001, two asterisks—p < 0.01, one asterisk—p < 0.05 (t-test, applying Benjamini–Hochberg p adjustment to correct for multiple testing).
Figure 11. Relative gene expression in flies infected with wMelCS112 and wMelPlus Wolbachia strains (lines Bi90wMelCS112 and Bi90wMelPlus, respectively) and uninfected flies of line Bi90T ± SEM. Asterisk indicates differences from the uninfected control. Three asterisks indicate p < 0.001, two asterisks—p < 0.01, one asterisk—p < 0.05 (t-test, applying Benjamini–Hochberg p adjustment to correct for multiple testing).
Ijms 24 17411 g011
Table 1. Number of RNA-seq reads mapped to the Drosophila melanogaster genome per sample.
Table 1. Number of RNA-seq reads mapped to the Drosophila melanogaster genome per sample.
SampleAll Reads (Forward + Reverse)AssignedUnassigned
Uninfected_129,820,84419,513,63410,307,210
Uninfected_219,028,51815,259,3473,769,171
Uninfected_315,927,16613,118,2102,808,956
Uninfected_423,642,11418,570,3935,071,721
wMelPlus_117,379,11714,243,0163,136,101
wMelPlus_215,325,06912,796,5522,528,517
wMelPlus_324,559,75018,973,6365,586,114
wMelCS112_122,813,21416,048,5716,764,643
wMelCS112_218,300,29815,087,7983,212,500
wMelCS112_322,668,14819,149,9093,518,239
Table 2. Comparison between females of two Drosophila melanogaster lines infected with wMelCS112 and wMelPlus Wolbachia strains by expression levels of annotated DEGs included in functional groups, in relation to the uninfected control group.
Table 2. Comparison between females of two Drosophila melanogaster lines infected with wMelCS112 and wMelPlus Wolbachia strains by expression levels of annotated DEGs included in functional groups, in relation to the uninfected control group.
Functional GroupRelative
Expression Level
StrainGene
Oxidation–reduction processUpregulatedBothrhi, Cyp6a13, Cyp6a14, TBCD, Crys, dgo
wMelCS112PPO1, Irc, Fatp3, oys, Fad2
wMelPlusPrx2540-1
DownregulatedBothCyp6d2, atk, Cap-D2, Cyp6a8, Cpn
wMelCS112ich, sha, sqa, nw, Zasp66, ninaG, Cyp309a2, rib, fj, tinc, hth
wMelPlusUro, PH4alphaEFB
Carbohydrate
metabolism
and transportation
UpregulatedBothMal-A2, Mal-A6, Mal-A1, Ugt35A1, Ugt36E1
wMelCS112Mal-A7, Amy-p, Hml, LManI, LManII, LManV, LManVI, Cht4, Ugt301D1
wMelPlusCht10, Gba1a
DownregulatedBothCht2, lectin-33A, Ugt35C1, Ugt302K1, fbp, Est-6
Transmembrane transportUpregulatedBothrtet, Atet, Zip99C
wMelCS112Fer1HCH, Ir40a, Tret1-2, NKCC, Ir76a
wMelPlusNtR, sut4, l(2)03659, COX7C
DownregulatedBothkcc
wMelCS112Irk2, ine, sand
wMelPlusMdr50, NaPi-T, Smvt
Alkaline
phosphatases
UpregulatedwMelCS112Alp2, Alp9, Alp10
wMelPlusPhu, Alp4
Response to stressUpregulatedBothBomBC2, BomT2
wMelPlusCrzR
DownregulatedBothTotA, TotC, TotX
Table 3. Comparison between females of two Drosophila melanogaster lines infected with wMelCS112 and wMelPlus Wolbachia strains by expression levels of annotated DEGs included in the “Embryogenesis” functional group in relation to the uninfected control group.
Table 3. Comparison between females of two Drosophila melanogaster lines infected with wMelCS112 and wMelPlus Wolbachia strains by expression levels of annotated DEGs included in the “Embryogenesis” functional group in relation to the uninfected control group.
Relative
Expression Level
StrainGeneSubgroup
UpregulatedBothrin, TBCD, dgo, gktEstablishment or maintenance of cell polarity
vls, rhi, rin, rasp, r2d2, simaOogenesis
CrysStructural constituent of eye lens
rhi, rin, rasp, r2d2, RhoGEF64C, dgo, HphEpithelium development
wMelCS112EgrCytokine receptor binding
lectin-37Da, lectin-37DbGalactose binding
oysLysophospholipid acyltransferase activity
InvadolysinMetalloendopeptidase activity
Crim, egrOther molecular function
wMelPlusTwdlL, TwdlJStructural constituent of chitin-based cuticle
Loxl1Cargo receptor activity
Lsp2Nutrient reservoir activity
stacCation binding
DownregulatedBothft, sns, stan, plum, otk2Cell adhesion molecule binding
Idgf3, Idgf2Chitinase activity
D, hth, nerfin-1, Oli, abDNA-binding transcription factor activity
blanksDouble-stranded RNA adenosine deaminase activity
sqaMyosin light chain kinase activity
spz6Signaling receptor binding
sloCalcium activated cation channel activity
HdcCarboxylyase activity
Zasp66Actinin binding
Ca-β, Cap-D2, beat-VII, atk, wkd, MFS14, cv-d, neo, sha, mas, nwOther molecular function
wMelCS112Irk2Inward rectifier potassium channel activity
bvesCAMP binding
ineSmall molecule sensor activity
pnt, ichDNA-binding transcription activator activity
TigIntegrin binding
Lectin-galC1Galactose binding
fjWnt-protein binding
Table 4. Comparison between females of two Drosophila melanogaster lines infected with wMelCS112 and wMelPlus Wolbachia strains by expression levels of annotated DEGs included in the “Proteolysis” functional group in relation to the uninfected control group.
Table 4. Comparison between females of two Drosophila melanogaster lines infected with wMelCS112 and wMelPlus Wolbachia strains by expression levels of annotated DEGs included in the “Proteolysis” functional group in relation to the uninfected control group.
Relative
Expression Level
StrainGeneSubgroup
UpregulatedBothphr6-4Deoxyribodipyrimidine photolyase activity
Vm32EStructural constituent of vitelline membrane
sbrProtein N-terminus binding
lokTau-protein kinase activity
BaceAspartic-type endopeptidase activity
raspPalmitoyltransferase activity
Jon44EEndopeptidase activity
tobiHydrolase activity, hydrolyzing O-glycosyl compounds
Obp99bOdorant binding
Vm32EOther molecular function
wMelCS112Nepl21, Nep6Metalloendopeptidase activity
Amy-pAlpha-amylase activity
egrTumor necrosis factor receptor binding
Agpat21-acylglycerol-3-phosphate O-acyltransferase activity
Myd88Toll binding
lectin-37DaGalactose binding
Phae1Catalytic activity, acting on a protein
DnaJ-HUnfolded protein binding
NimC1, Cul6, Agpat2, BomBc1, HmlOther molecular function
wMelPlusGasp, obst-A, Cda4, vermChitin binding
epsilonTry, Jon66Ci, Jon66Cii, Jon99Fi, Nepl11, PsaPeptidase activity
Pdk, Gasp, obst-A, bves, Cda4, verm, hllCarbohydrate derivative binding
Lsp2Nutrient reservoir activity
yellow-g2, yellow-gIsomerase activity
Down
regulated
Bothneo, Muc12Ea, Muc30E, Muc68D,Extracellular matrix structural constituent
LysP, mas, Nepl3, Jhedup, Jon65Aii, mag, Sirt6, yip7, PGRP-SC2, PGRP-SC1a, Cda5, Send2Hydrolase activity
Npc2d, Npc2fSteroid binding
Lectin-galC1Galactose binding
tnTranslation repressor activity
Cp16, Clk, Ag5r2, Obp56hOther molecular function
wMelCS112Pdk, Gasp, Vajk1, obst-A, bves, Cda4, verm, hllCarbohydrate derivative binding
spz6Toll binding
Jon66Cii, Nepl11Endopeptidase activity
Obp83a, TpnC73F, ImpE2Other molecular function
wMelPlusSer8, Jon65Ai, Jon99Fii, Sp7, lambdaTry, omeSerine-type peptidase activity
CAH2Carbonate dehydratase activity
fonOther molecular function
Table 5. Primer sequences used for qRT-PCR.
Table 5. Primer sequences used for qRT-PCR.
Gene NameForwardReverse
TotAAATTCTTCAACTGCTCTTATGTGCTCAGCAATTCTAAGGTTGTCAGC
TotCCAGTTTGTCTTAAACCAGTGCTCCAGATTCCCTTTCCTCGTCAG
Ugt35CCAGACGAGATGATGAGTTTGCTACCAGATCGAAAGTTTCTCCC
CrzRAATCCGGACAAAAGGCTGGGAGGTGGAAGGCACCGTAGAT
lectin-33AGAGTCGGAAACAAGTGCTACCCGTGGTTGTGAGGAGTTTGTC
β-TubulinTGTCGCGTGTGAAACACTTCAGCAGGCGTTTCCAATCTG
Actin 5CGCGCCCTTACTCTTTCACCAATGTCACGGACGATTTCACG
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Gruntenko, N.E.; Deryuzhenko, M.A.; Andreenkova, O.V.; Shishkina, O.D.; Bobrovskikh, M.A.; Shatskaya, N.V.; Vasiliev, G.V. Drosophila melanogaster Transcriptome Response to Different Wolbachia Strains. Int. J. Mol. Sci. 2023, 24, 17411. https://doi.org/10.3390/ijms242417411

AMA Style

Gruntenko NE, Deryuzhenko MA, Andreenkova OV, Shishkina OD, Bobrovskikh MA, Shatskaya NV, Vasiliev GV. Drosophila melanogaster Transcriptome Response to Different Wolbachia Strains. International Journal of Molecular Sciences. 2023; 24(24):17411. https://doi.org/10.3390/ijms242417411

Chicago/Turabian Style

Gruntenko, Nataly E., Maksim A. Deryuzhenko, Olga V. Andreenkova, Olga D. Shishkina, Margarita A. Bobrovskikh, Natalja V. Shatskaya, and Gennady V. Vasiliev. 2023. "Drosophila melanogaster Transcriptome Response to Different Wolbachia Strains" International Journal of Molecular Sciences 24, no. 24: 17411. https://doi.org/10.3390/ijms242417411

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop