Hostname: page-component-7bb8b95d7b-5mhkq Total loading time: 0 Render date: 2024-09-28T23:17:13.591Z Has data issue: false hasContentIssue false

Analysis of the immune transcriptome of the invasive pest spotted wing drosophila infected by Steinernema carpocapsae

Published online by Cambridge University Press:  27 September 2024

A. Garriga*
Affiliation:
Departament de Biologia Animal, Biologia Vegetal i Ecologia, Facultat de Biociències, Universitat Autònoma de Barcelona, Bellaterra, Spain Centro de Biotecnologia dos Açores, Faculdade de Ciências e Tecnologia, Universidade dos Açores, Ponta Delgada, Portugal
D. Toubarro
Affiliation:
Centro de Biotecnologia dos Açores, Faculdade de Ciências e Tecnologia, Universidade dos Açores, Ponta Delgada, Portugal
A. Morton
Affiliation:
Departament de Biologia Animal, Biologia Vegetal i Ecologia, Facultat de Biociències, Universitat Autònoma de Barcelona, Bellaterra, Spain
N. Simões
Affiliation:
Centro de Biotecnologia dos Açores, Faculdade de Ciências e Tecnologia, Universidade dos Açores, Ponta Delgada, Portugal
F. García-del-Pino
Affiliation:
Departament de Biologia Animal, Biologia Vegetal i Ecologia, Facultat de Biociències, Universitat Autònoma de Barcelona, Bellaterra, Spain
*
Corresponding author: A. Garriga; Email: [email protected]
Rights & Permissions [Opens in a new window]

Abstract

Drosophila suzukii is a pest of global concern due to its great impact on several crops. The entomopathogenic nematode Steinernema carpocapsae was highly virulent to the larvae of the fly although some immune mechanisms were triggered along the infection course. Thus, to understand the gene activation profile we performed a comparative transcriptome of D. suzukii larvae infected with S. carpocapsae and Xenorhabdus nematophila to map the differentially expressed genes involved in the defence response. The analysis exposed the induction of genes involved in the humoral response such as the antimicrobial peptides and pattern-recognition receptors while there was a suppression of the cellular defence. Besides, genes involved in melanisation, and clot formation were downregulated hindering the encapsulation response and wound healing. After the infection, larvae were in a stress condition with an enrichment of metabolic and transport functionalities. Concerning the stress response, we observed variations of the heat-shock proteins, detoxification, and peroxidase enzymes. These findings set a genetical comprehensive knowledge of the host-pathogen relation of D. suzukii challenged with S. carpocapsae which could support further comparative studies with entomopathogenic nematodes.

Type
Research Paper
Creative Commons
Creative Common License - CCCreative Common License - BYCreative Common License - NCCreative Common License - SA
This is an Open Access article, distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike licence (http://creativecommons.org/licenses/by-nc-sa/4.0), which permits non-commercial re-use, distribution, and reproduction in any medium, provided the same Creative Commons licence is used to distribute the re-used or adapted article and the original article is properly cited. The written permission of Cambridge University Press must be obtained prior to any commercial use.
Copyright
Copyright © The Author(s), 2024. Published by Cambridge University Press

Introduction

Drosophila suzukii (Diptera: Drosophilidae), widely known as Spotted Wing Drosophila, is a globally invasive pest that significantly impacts soft-skinned and stone fruits such as berries, cherries, and strawberries (Lee et al., Reference Lee, Bruck, Curry, Edwards, Haviland, Van Steenwyk and Yorgey2011; Walsh et al., Reference Walsh, Bolda, Goodhue, Dreves, Lee, Bruck, Walton, O'Neal and Zalom2011). Initially introduced in America, it rapidly spread across Europe, causing great economic losses due to its polyphagous nature (Asplen et al., Reference Asplen, Anfora, Biondi, Choi, Chu, Daane, Gibert, Gutierrez, Hoelmer, Hutchison, Isaacs, Jiang, Kárpáti, Kimura, Pascual, Philips, Plantamp, Ponti, Vetek, Vogt, Walton, Yu, Zappala and Desneux2015). Furthermore, effective control measures have been challenging due to the quick generation span of the fly which requires frequent interventions. Gress and Zalom (Reference Gress and Zalom2019) reported resistance of D. suzukii to the chemical pesticide Spinosad, indicative of a potent detoxification response mechanism. Therefore, the prospection of biological control agents progressed by pointing to parasitoids and entomopathogenic nematodes (EPNs) as promising candidates to control the pest (Wang et al., Reference Wang, Daane, Hoelmer, Lee and Mello Garcia2021). The results with parasitoids varied, with pupal parasitoids being successful, while D. suzukii larvae mounted a strong immune response to encapsulate parasitoid eggs (Kacsoh and Schlenke, Reference Kacsoh and Schlenke2012). In contrast, the EPN Steinernema carpocapsae (Steinernema: Panagrolaimida) was highly effective against both larvae and adults of D. suzukii under laboratory conditions (Garriga et al., Reference Garriga, Morton and Garcia-del-Pino2018, Reference Garriga, Morton, Ribes and Garcia-del-Pino2020b). Given these differences in larval responses to parasitoid eggs and EPNs, our goal was to prospect the immune response elicited by the nematode-bacterial complex in D. suzukii.

EPNs belonging to the families Steinernematidae and Heterorhabditidae are obligate parasites of a wide range of insects used for the biological control of pests (Woodring and Kaya, Reference Woodring and Kaya1988). The infection process by EPNs is intricate due to the symbiosis complex of nematode and the bacteria Xenorhabdus nematophila, which impacts many pathways from the insect immune response (Peña et al., Reference Peña, Carrillo and Hallem2015). The infective process of any pathogen involves a wide range of genes tangled together, from those restricting metabolic pathways to those activating stress and defence mechanisms, to focus resources on overcoming the infection. It is therefore expected to observe an activation of the immune-promotor genes shortly after infection with the triggering of the pattern recognitions proteins that identify the foreign body as non-self (Lemaitre and Hoffmann, Reference Lemaitre and Hoffmann2007). Combating the immune response is crucial for the success of the EPNs which evolved with a dual purpose. Firstly, to evade host recognition through camouflage and evasive strategies (Brivio et al., Reference Brivio, Toscano, De Pasquale, Barbaro, Giovannardi, Finzi and Mastore2018; Brivio and Mastore, Reference Brivio and Mastore2020). Secondly, to counter the immune response with the secretions produced by both nematode and bacteria (Toubarro et al., Reference Toubarro, Avila, Montiel and Simões2013; Eliáš et al., Reference Eliáš, Hurychová, Toubarro, Frias, Kunc, Dobes, Simoes and Hyrsl2020; Jones et al., Reference Jones, Tafesh-Edwards, Kenney, Toubarro, Simoes and Eleftherianos2022). In the case of D. suzukii, the fly was unable to encapsulate the nematode as seen in previous experiments which evaluated the physiological aspects of this infective process (Garriga et al., Reference Garriga, Mastore, Morton, Garcia-del-Pino and Brivio2020a). During these experiments, the cellular response was either ineffective or not triggered by the presence of the nematodes, resulting in a high success rate of nematode infection. Nevertheless, the humoral response was activated by the presence of the symbiont bacteria X. nematophila in the haemolymph but was unable to block the progress of the nematode-bacterial infection.

The biological assessment and differential expression of a few genes provided an initial understanding of the infection and defence responses, although many gaps remained uncovered (Garriga et al., Reference Garriga, Toubarro, Simões, Morton and Garcia-del-Pino2023). Hence, the utilisation of mRNA sequencing (RNA-Seq) technology provides a wider and more comprehensive perspective of the infection process. This technique allows for a deeper understanding of the infection dynamics, unveiling intricate physiological details such as the effects in metabolic processes and stimulus-response pathways of the insects as seen in nematode-bacterial infections in Drosophila melanogaster (Diptera: Drosophilidae) larvae (Arefin et al., Reference Arefin, Kucerova, Dobes, Markus, Strand, Wang, Hyrsl, Zurovec and Theopold2014; Castillo et al., Reference Castillo, Creasy, Kumari, Shetty, Shokal, Tallon and Eleftherianos2015). The sequencing of transcripts further facilitates the identification of evolutionary distinctions when compared to established species, revealing adaptive strategies that potentially enhance the adaptability and proliferation of the pest (Mérel et al., Reference Mérel, Gibert, Buch, Rodriguez-Rada, Estoup, Gautier, Fablet, Boulesteix and Vieira2021). Due to the advances in transcriptome sequencing and gene annotation, insects such as Spodoptera frugiperda (Lepidoptera: Noctuidae) and Holotrichia parallela (Coleoptera: Scarabaeidae) can be analysed with this technique to directly study the infective process of EPNs on the insect pest of interest (Huot et al., Reference Huot, George, Girard, Severac, Negre and Duvic2019; Li et al., Reference Li, Qin, Feng, Li, Li, Nyamwasa, Cao, Ruan, Li and Yin2021). Previous studies have examined the transcriptome of D. suzukii, focusing on traits like olfactory receptors, reproduction, detoxification, and diapause (Shearer et al., Reference Shearer, West, Walton, Brown, Svetec and Chiu2016; Mishra et al., Reference Mishra, Chiu, Hua, Tawari, Adang and Sial2018; Schwanitz et al., Reference Schwanitz, Polashock, Stockton, Rodriguez-Saona, Sotomayor, Loeb and Hawkings2022; Xing et al., Reference Xing, Deng, Wen and Peng2022). These studies have contributed to understanding the evolutionary dynamics of the fly and assisted in predicting and managing its invasion, although none have focused on the immune response to pathogens.

In this perspective, our objective was to use a comparative transcriptomic analysis to elucidate functional changes and gene regulations in D. suzukii larvae infected with the nematode complex S. carpocapsae – X. nematophila. This data could provide a better understanding of host-pathogen interactions and contribute to the development of targeted future pest management strategies.

Material and methods

Insects and EPNs culture

For the study, we used third-instar larvae of D. suzukii produced in a laboratory culture established in 2017 from wild specimens. Insects were reared on a modified drosophila diet (Garriga et al., Reference Garriga, Morton and Garcia-del-Pino2018) at 25°C with a 12:12 h photoperiod. The infection experiment was conducted under these climatical conditions.

The assay was performed with S. carpocapsae (strain B14) isolated from urban garden soil in Barcelona, Catalonia (NE Spain). The native bacteria were replaced by the X. nematophila Green Fluorescent Protein (GFP-labelled strain F1D3) provided by the laboratory of Prof. Givaudan (University of Montpellier, France) (Garriga et al., Reference Garriga, Mastore, Morton, Garcia-del-Pino and Brivio2020a). Nematodes were reared in the late instar G. mellonella larva at 25°C (Woodring and Kaya, Reference Woodring and Kaya1988). Once the IJs emerged from the insect, IJs were stored in sterile tap water (STW) at 9°C for a maximum of two weeks. Before use, the nematodes were acclimatised at room temperature for 3 h to ensure maximum activation during the assay.

Infection assay

The infection assay was performed following the methodology described by Garriga et al. (Reference Garriga, Toubarro, Simões, Morton and Garcia-del-Pino2023). Larvae were exposed to 300 IJs in 96-well plates filled with sterile filter paper for 2 h. After the exposition, we rinsed larvae thoroughly with STW and transferred to a new Petri dish with filter paper and a cube of drosophila diet to allow the infection to develop for 2 more hours. After this incubation period, larvae were subjected to a final rinse with STW before being transferred individually to a 1.5 ml tube containing 20 μl of RNAlater and immediately frozen at −80°C. The same procedure of manipulation and rinsing steps were conducted with control larvae which only received STW as exposure treatment. Drosophila suzukii larvae were dissected individually in RNAlater to verify the nematode entry by observation of IJs (mean entry of 6.68 ± 4.45 IJs per larvae). Only larvae with confirmed nematode presence were used for RNA extraction. These larvae were pooled into groups of five per treatment for the RNA extraction, and three biological repetitions were performed for both control and infected larvae.

mRNA collection and sequencing

RNA isolation was performed with TRIzol Reagent combined with Invitrogen PureLink RNA Mini Kit (ThermoFisher), following the manufacturer's instructions. Purified RNA was kept at −80°C until the sequencing step. The quality and quantity of RNA were assessed with Nanodrop and the RNA integrity confirmed with Bioanalyzer Samples were sequenced with the Illumina NovaSeq platform using 150 bp paired-end sequence reads with the library for stranded mRNA preparation kit. The reads were trimmed to remove low coverage and adaptors. The quality of the reads was assessed with Fast-QC before continuing with the bioinformatic process (Andrews, Reference Andrews2010).

Bioinformatic and statistical procedure

The reads were assembled with the D. suzukii genome of reference GCF_013340165.1 from the NCBI database. The assembly was done with the software Bowtie2 altogether with the software RSEM to make the counts of the transcripts (Li and Dewey, Reference Li and Dewey2011). Differential analysis was conducted using the DESeq2 package to obtain the significant differentially expressed genes (DEG) with a cutoff of False Discovery Rate (FDR) less than 0.1 and an absolute Log2 Fold-Change of 0.5 (Love et al., Reference Love, Huber and Anders2014). The functional annotation was done using the Sma3s software with the Uniref90 database for the assignment of the Gene Ontology (GO) Terms (Casimiro-Soriguer et al., Reference Casimiro-Soriguer, Muñoz-Mérida and Pérez-Pulido2017). The pathway information was obtained from the Kyoto Encyclopaedia of Genes and Genomes (KEGG) through the EggNOG platform as well as the KASS platform (Moriya et al., Reference Moriya, Itoh, Okuda, Yoshizawa and Kanehisa2007; Cantalapiedra et al., Reference Cantalapiedra, Hernández-Plaza, Letunic, Bork and Huerta-Cepas2021). We discriminated the enriched terms from the DEG compared to the complete set of mRNA using the ClusterProfiler and TopGO packages (Wu et al., Reference Wu, Hu, Xu, Chen, Chen, Guo, Dai, Feng, Zhou, Tang, Zhan, Fu, Liu, Bo and Yu2021; Alexa and Rahnenfuhrer, Reference Alexa and Rahnenfuhrer2023). A manual curation helped by the GO terms was used to filter the genes of immune defence and stimulus-response for a closer assessment of these pathways. The visualisation of results was done with the ggplot2 package. All statistical analysis packages were conducted with RStudio (R Core Team, 2017).

Results

Sequencing results and differential analysis

Infection with EPN was observed in 75% of the larvae dissected in the present test. We sequenced the complete transcriptome of infected D. suzukii larvae with S. carpocapsae B14 and the symbiont bacteria X. nematophila F1D3. From the sequencing process, a mean number of 71 million reads was obtained per sample once the adaptors and low-quality were trimmed (table 1). The sequenced reads can be found in the NCBI SRA database with the code PRJNA910932. The mapped reads reached a percentage of 65 to 74% across samples with a total of 15,567 identified genes through the assembly with the D. suzukii reference genome. The transcriptome of D. suzukii was functionally annotated for GO terms reaching an identification of 12,806 transcripts, while 8149 transcripts were identified using the KEGG database.

Table 1. Summary statistics of transcriptome sequencing

In the principal component analysis (PCA), the PC2 explains 31% of the variance which include transcripts that separate the samples of nematode-infected larvae from the control ones (fig. S1). From the identified sequences, 242 transcripts were differentially expressed genes (DEG) comparing control and nematode-infected larvae. From these genes, 104 were upregulated and 138 were downregulated (fig. 1, table S1). Among the DEG, our analysis identified 40 genes annotated in the defence response category while 50 genes belong to the stress response system. The metabolism of larvae presented many alterations (93 DEG) mainly related to secondary metabolism as well as significant variations of the transmembrane transportation (21 DEG). The genes of immune defence and stress altered after the EPNs infection will be discussed in detail further below.

Figure 1. Volcano plot representation of the differentially expressed genes comparing the transcriptome of infected and control larvae.

Functional analysis

The functional analysis displayed enrichment of the GO terms corresponding to the categories Biological Process (BP), Molecular Function (MF), and Cell Component (CC) (fig. 2). The most enriched functionalities from the BP were related to the defence response and immune reaction. Among those, we highlight the enrichment of the antibacterial response, humoral immune mechanisms, and inflammatory response in line with an infection course. A general stress activation was also observed by the enrichment of the processes related to responses to other organisms, cold, fungi, and biotic stimuli. Moreover, we observed the involvement of metabolic functions such as the diol process, the carboxylic acid, and the organic acid biosynthetic. Regarding the MF, the more enriched terms were the structural constituent of cuticle and the iron ion binding, which are categories characteristically tight to pathogen infection. We also detected an enrichment of the activity aldo-keto reductase (NAPD), oxidoreductase, monooxygenase, and catalytic categories. The alteration of these enzymatic processes is linked to the metabolic swift due to the stress condition of larvae. In the cell component category (CC), the extracellular space was enriched indicating the increase of the transport components across the cellular space and haemolymph can be related to humoral and detoxification processes.

Figure 2. Significantly enriched terms of Gene Ontology (GO) after the analysis of the DEG using TopGO, considering the categories: biological process (BP), Molecular Function (MF), and Cell Component (CC).

Regarding the pathway KEGG categories (level 2) that were altered after the infection of nematodes were related to metabolism and ABC transporters, with the enrichment of the biosynthesis of secondary metabolites, and the metabolism of ether lipid, sphingolipid, ascorbate and aldarate, porphyrin, steroid biosynthesis, and retinol (fig. 3A). The KEGG analysis at the gene level (level 3) further revealed that the genes Cecropin and Diptericin were strongly enriched together with the immune-related enzyme Aminopeptidase N (fig. 3B). Genes coding for metabolic enzymes such as ceramide galactosyltransferase, glucuronosyltransferase, and choline dehydrogenase were also enriched. The enrichment of the gene DnaJ homologue evidences the implication of translation processes and heat shock proteins.

Figure 3. Significantly enriched terms (x-axis) using the KEGG database and the transcript count (y-axis) after the analysis of the DEG, considering (A) the level 2 pathways and (B) the level 3 genes.

DEG of immune defence

The immune defence is a complex system that involves many branched processes, so the basal GO term immune system process (GO 0002376) was our base point to analyse the annotated transcripts as such. After the filtering of low counts, 1024 transcripts were included in this GO term with 40 significant DEG (table 2). As pointed out by the enrichment of the defence process, the most upregulated genes after the nematode infection belong to the Imd pathway. Among them, there were two PGRP receptors homologues to PGRP-SB and -SD of D. melanogaster which were significantly upregulated. Furthermore, the activation reached the AMP effector genes in at least seven different genes of the Imd pathway (AttB, CecB, CecC, Cec1, AttB, DptB, Dpt) with a fold-change greater than 2. In the Toll pathway, we identified the upregulation of the two AMP effector genes Drosocin and Metchnikowin after the nematode infection. Moreover, there were upregulated genes with signalling roles or humoral compounds from these pathways' cascades such as edin (XM_017067854), slif (XM_017071440), IDGF (XM_017082031), sno (XM_017089152), and ficolin-1 (XM_017072621).

Table 2. List of significant DEG associated with immune defence responses upon S. carpocapsae infection, with the mRNA identification (ID RNA), the protein identification (ID Protein), and the gene name obtained during the annotation step

While the antimicrobial pathways were activated up to the effector genes, other immune responses more specialised in cellular defence were suppressed. This is the case of the effector gene TurandotB (XM_017086060) from Jak-STAT strongly downregulated and the negative feedback socs4 (suppressor cytokine XM_017090197) which was upregulated. Genes belonging to the PPO pathway did not present a significant variation and the serine protease Sp7 (XM_017073757) that activates the melanisation response was downregulated. Additionally, the transglutaminase gene (XM_017087002) which plays an essential role as a clotting factor was also downregulated. Besides, the downregulation of two chitinase genes (XM_036818866 and XM_036817002) associated with wound healing could indicate a compromised capacity for healing tissues after nematode infection.

DEG of the stress response

The transcripts related to stress and stimulus-response represented a large portion of the 242 DEG indicating the stress condition of the larvae after the infection (table S2). Among these, we highlight the significant upregulation of the heat-shock genes hsp22 and mcm5-hsp70 (XM_017077979 and XM_017071045). Furthermore, several genes of multidrug resistance proteins (XM_017070631 and XM_036818694) and the drug transmembrane transport Mdr49 (XM_017085672) were upregulated, which also explains the functional enrichment of the ABC transporters. However, the detoxification genes related to UDP-glycosyltransferase activity (XM_017075039, XM_036819290, and XM_036816950) were downregulated. Besides, the downregulation of two peroxidase genes and oxidoreductase limited the operation of the ROS mechanism. Concerning cellular regulation, the genes NimCs and the gene encoding for the protein Croquemort were downregulated after the nematode infection. These genes are implicated in the process of cell death, phagocytosis, and haemocyte regulation.

Discussion

EPNs undergo a dynamic relationship with insects, leading to a constant coevolution of defensive mechanisms to combat infection. The S. carpocapsae infection induced a distinct activation of the immune mechanisms in D. suzukii larvae, based on our previous physiological assessment (Garriga et al., Reference Garriga, Mastore, Morton, Garcia-del-Pino and Brivio2020a). Thus, the results of the current work with transcriptomic sequencing provide a wider view and help to uncover the gaps concerning the genes regulating these physiological mechanisms.

Through the analysis of the comparative transcriptome, we identified a total of 242 genes induced after infection. Our results suggest that infected larvae reduce metabolic processes to focus resources on confronting the infection. Nevertheless, only a portion of the immune responses was triggered, primarily activating antimicrobial defence relying on humoral components, while concurrently restraining other mechanisms like cellular defence. These defence actions of D. suzukii, pointed out by the RNA-Seq, are in concordance with the results from the q-PCR technique, which serve as validation of the current work (Garriga et al., Reference Garriga, Toubarro, Simões, Morton and Garcia-del-Pino2023). There was recognition of the pathogen by the Imd PGRP receptors-SB, -SD, and -LC while PGRP-LF was not regulated in either q-PCR or RNASeq results. In accordance with our findings, D. melanogaster infected with S. carpocapsae showed an upregulation of only some PGRP genes (Yadav et al., Reference Yadav, Daugherty, Shetty and Eleftherianos2017). The upregulation of pattern recognition genes conveyed the enrichment of the extracellular matrix region, also reported in infected D. melanogaster (Arefin et al., Reference Arefin, Kucerova, Dobes, Markus, Strand, Wang, Hyrsl, Zurovec and Theopold2014). The AMPs Cecropin, Dipericin, and Attacin were upregulated, which indicates the detection of nematodes or at least their symbiotic bacteria, in agreement with q-PCR results within 4 h after infection (Garriga et al., Reference Garriga, Toubarro, Simões, Morton and Garcia-del-Pino2023). The Imd pathway upregulation is typically associated with nematode-bacterial complexes, as when only axenic nematodes infect a fly, a lower activation of the AMP gene was achieved (Peña et al., Reference Peña, Carrillo and Hallem2015; Yadav et al., Reference Yadav, Daugherty, Shetty and Eleftherianos2017). Concerning the Toll pathway, Defensin was not significantly altered at 4 h post-infection in both analyses, although q-PCR results at 14 h showed a huge increase, suggesting later gene activation (Garriga et al., Reference Garriga, Toubarro, Simões, Morton and Garcia-del-Pino2023). Transcriptome data revealed the upregulation of the Metchnikowin and Drosocin genes, which belong to the Toll pathway. Previous works reported a large upregulation of AMPs in this pathway during EPNs infection course (Castillo et al., Reference Castillo, Creasy, Kumari, Shetty, Shokal, Tallon and Eleftherianos2015; Huot et al., Reference Huot, George, Girard, Severac, Negre and Duvic2019). Indeed, the Toll pathway is associated not only with infection of Gram-positive bacteria but also with metazoan pathogens such as nematodes (Shan et al., Reference Shan, Wang, Bhattarai and Jiang2023). These results may also hint the presence of additional bacteria in the insect haemolymph besides X. nematophila, which could originate from the microbiome of the nematode called pathobiome (Ogier et al., Reference Ogier, Pagès, Frayssinet and Gaudriault2020).

The transcriptomic data confirmed previous physiological observations that the cellular response is constrained during infection. The cell metabolic enzyme AMP deaminase was downregulated, as well as two defensive genes encoding chitinase enzymes, which are crucial for wound healing during nematode infection (Kucerova et al., Reference Kucerova, Broz, Arefin, Maaroufi, Hurychova, Strand, Zurovec and Theopold2016). Furthermore, two apoptotic genes were downregulated in response to nematode entry, as previously reported in S. frugiperda (Huot et al., Reference Huot, George, Girard, Severac, Negre and Duvic2019). There was a strong downregulation of Turandot B, which could be correlated with the TotC expression observed by q-PCR which suggests a lack of activation of the Jak/STAT pathway (Garriga et al., Reference Garriga, Toubarro, Simões, Morton and Garcia-del-Pino2023). This observation aligns with findings in D. melanogaster where infection with S. carpocapsae also exhibited lower induction of the JNK and Jak/STAT pathways compared to the humoral pathways (Yadav et al., Reference Yadav, Daugherty, Shetty and Eleftherianos2017). However, in contrast to D. suzukii, the fruit fly exhibited higher activation of the Turandot family (Castillo et al., Reference Castillo, Shokal and Eleftherianos2013; Yadav et al., Reference Yadav, Frazer, Banga, Pruitt, Harsh, Jaenike and Eleftherianos2018). In the biological tests performed with D. suzukii haemocytes, the cells were unable to detect S. carpocapsae and adhere to the nematode surface (Garriga et al., Reference Garriga, Mastore, Morton, Garcia-del-Pino and Brivio2020a). This evasive strategy, based on the composition of the nematode's cuticle, was previously described in G. mellonella and added to the secretion of proteases from S. carpocapsae facilitate the evasion of the cellular encapsulation (Mastore and Brivio, Reference Mastore and Brivio2008; Toubarro et al., Reference Toubarro, Avila, Montiel and Simões2013). Another fundamental aspect of the cellular response involves the processes of melanisation and haemolymph coagulation, which should be rapidly activated upon infection. However, the genes encoding serine proteases and transglutaminase, which promote melanisation and clot formation (Lindgren et al., Reference Lindgren, Riazi, Lesch, Wilhelmsson, Theopold and Dushay2008; Shan et al., Reference Shan, Wang, Bhattarai and Jiang2023), were downregulated in our EPN-infected larvae. The absence of PPO genes in the DEG supports the lack of differential regulation observed by q-PCR amplification of the genes PPO1 and PPO2 in D. suzukii. The PPO pathway is usually associated with EPN infection due to the formation of melanin clots and contribution to the encapsulation process, although PPO activation was restricted in other nematode–insect infections (Huot et al., Reference Huot, George, Girard, Severac, Negre and Duvic2019; Dziedziech et al., Reference Dziedziech, Shivankar and Theopold2020). In D. melanogaster, it was also hypothesised that wound healing involves post-transcriptional products already present in the haemolymph, with no gene expression needed during infection (Arefin et al., Reference Arefin, Kucerova, Dobes, Markus, Strand, Wang, Hyrsl, Zurovec and Theopold2014).

Following S. carpocapsae infection, the induction of stress status in D. suzukii larvae was apparent, with upregulation of several stress indicators and the functional enrichment of stimulus and stress response categories. This induction was also reported in D. melanogaster adults infected with H. bacteriophora (Yadav et al., Reference Yadav, Daugherty, Shetty and Eleftherianos2017). A high upregulation was confirmed for two Hsp genes, crucial for stress response mechanisms (Sorensen et al., Reference Sorensen, Nielsen, Kruhoffer, Justesen and Loeschcke2005). These genes indicate cellular stress and modulate the inflammatory process, and were already reported to be upregulated during EPN infection (Yadav et al., Reference Yadav, Daugherty, Shetty and Eleftherianos2017; Huot et al., Reference Huot, George, Girard, Severac, Negre and Duvic2019). The activation of enzymes associated with lipid metabolism could also indicate internal tissue damage caused by nematodes and an intent to repair it (McIntire et al., Reference McIntire, Yeretssian and Saleh2009). In contrast, infected D. suzukii exhibited downregulation of peroxidase and oxidoreductase genes, which contribute to antimicrobial defence by generating reactive oxygen species (ROS). Concerning detoxification mechanisms, we detected that while three annotated genes as UDP-glycosyltransferase were downregulated, other genes were strongly upregulated such as the multidrug resistance genes from the ABC transporter superfamily which acts as efflux for toxins from cells. In addition, D. suzukii presented alterations in iron binding functionalities, an indicator of pathogen presence, as ferric compounds are crucial resource during infections (Akinbosede et al., Reference Akinbosede, Chizea and Hare2022; Liu et al., Reference Liu, Zhu, Guo, Zhu, Huang, Cao, Yu, Liu and Xu2022). The upregulation of these genes could result from nematode and bacterial secretions and may be part of the stress response mechanisms linked to specific tissues, as previously reported in S. frugiperda (Huot et al., Reference Huot, George, Girard, Severac, Negre and Duvic2019). We observed a functional alteration of the DNA translation process in D. suzukii, which were also pointed out in D. melanogaster infected with the nematode H. bacteriophora (Castillo et al., Reference Castillo, Creasy, Kumari, Shetty, Shokal, Tallon and Eleftherianos2015). These results may indicate the nematodes’ capacity to disrupt gene transcription within the fly, although further experiments should be done to explore this ability.

Transcriptome data represents only a snapshot of the infection course, and a previous or subsequent activation of the cellular response could still occur. For this reason, biological assessments are important to evidence that D. suzukii haemocytes are unable to recognise and encapsulate S. carpocapsae (Garriga et al., Reference Garriga, Mastore, Morton, Garcia-del-Pino and Brivio2020a). The implications of this type of study extend beyond the description of affected functions and implicated genes. With enhanced knowledge of transcriptomic alternations, we can unravel novel pathways or regulatory networks affected by the nematode-bacterial complex, contributing to our understanding the host-pathogen interactions. Moreover, this genetic knowledge provides hints for designing future biological control tools such as genetic targeting using interference RNA or selecting EPNs to surpass the immune system of the insect.

In conclusion, the transcriptomic analysis aligned with the physiological assessments, as D. suzukii infected with S. carpocapsae exhibited distinct immune activation, focusing on humoral components, while restraining cellular defence. Stress responses, including upregulation of heat shock protein, and alterations in detoxification and DNA translation processes, indicated broader physiological impacts of the infection. These findings emphasise the importance of understanding host-pathogen interactions, as despite the activation detected in the immune system, EPNs succeeded in the infection. These results contribute to a better understanding of immune mechanisms targeted by EPNs and provide valuable insights for designing effective management strategies for this agricultural pest.

Supplementary material

The supplementary material for this article can be found at https://doi.org/10.1017/S0007485324000543.

Data

The datasets of this study can be found in the NCBI repository SRA database under the Project PRJNA910932 and BioSamples SRR22675644, SRR22675643, SRR22675642, SRR22675641, SRR22675640, SRR22675639.

Acknowledgements

We appreciate the support of Antonio Muñoz Mérida (CiBIO InBIO) for the bioinformatic procedures, and Miguel Teruel for the collaboration on the graphical representation.

Author contributions

Conceptualisation: F. G. P., A. M., and N. S. Experimental design: D. T., and A. G. Performed experiments and analysed data: A. G. Writing original draft: A. G. Writing – review and editing: F. G. P., N. S., D. T., A. M., and A. G. Funding acquisition: F. G. P. and N. S. All authors have read and agreed to the published version of the manuscript.

Financial support

The research presented in this paper was supported by FEDER/Ministerio de Ciencia, Innovación y Universidades – Agencia Estatal de Investigación/Project (AGL2017-86770-R) and the project PTDC/BAA/30609/2017 of FCT.

Competing interests

None.

Ethical standards

All applicable international, national, and/or institutional guidelines for the care and use of animals were followed.

References

Akinbosede, D, Chizea, R and Hare, SA (2022) Pirates of the haemoglobin. Microbial Cell 9, 84102. https://doi.org/10.15698/MIC2022.04.775.CrossRefGoogle ScholarPubMed
Alexa, A and Rahnenfuhrer, J (2023) topGO: enrichment analysis for gene ontology. R Packag. version 2.52.0.Google Scholar
Andrews, S (2010) FastQC: a quality control tool for high throughput sequence data [Online]. Available at http://www.bioinformatics.babraham.ac.uk/projects/.Google Scholar
Arefin, B, Kucerova, L, Dobes, P, Markus, R, Strand, H, Wang, Z, Hyrsl, P, Zurovec, M and Theopold, U (2014) Genome-wide transcriptional analysis of drosophila larvae infected by entomopathogenic nematodes shows involvement of complement, recognition and extracellular matrix proteins. Journal of Innate Immunity 6, 192204. https://doi.org/10.1159/000353734.CrossRefGoogle ScholarPubMed
Asplen, MK, Anfora, G, Biondi, A, Choi, DS, Chu, D, Daane, KM, Gibert, P, Gutierrez, AP, Hoelmer, KA, Hutchison, WD, Isaacs, R, Jiang, ZL, Kárpáti, Z, Kimura, MT, Pascual, M, Philips, CR, Plantamp, C, Ponti, L, Vetek, G, Vogt, H, Walton, VM, Yu, Y, Zappala, L and Desneux, N (2015) Invasion biology of spotted wing Drosophila (Drosophila suzukii): a global perspective and future priorities. Journal of Pest Science 88, 469494. http://dx.doi.org/10.1007/s10340-015-0681-z.CrossRefGoogle Scholar
Brivio, MF and Mastore, M (2020) When appearance misleads: the role of the entomopathogen surface in the relationship with its host. Insects 11, 124. https://doi.org/10.3390/insects11060387.CrossRefGoogle ScholarPubMed
Brivio, MF, Toscano, A, De Pasquale, SM, Barbaro, AL, Giovannardi, S, Finzi, G and Mastore, M (2018) Surface protein components from entomopathogenic nematodes and their symbiotic bacteria: effects on immune responses of the greater wax moth, Galleria mellonella (Lepidoptera: Pyralidae). Pest Management Science 74, 20892099. https://doi.org/10.1002/ps.4905.CrossRefGoogle Scholar
Cantalapiedra, CP, Hernández-Plaza, A, Letunic, I, Bork, P and Huerta-Cepas, J (2021) eggNOG-mapper v2: functional annotation, orthology assignments, and domain prediction at the Metagenomic scale. Molecular Biology and Evolution 38, 58255829. https://doi.org/10.1093/molbev/msab293.CrossRefGoogle ScholarPubMed
Casimiro-Soriguer, CS, Muñoz-Mérida, A and Pérez-Pulido, AJ (2017) Sma3s: a universal tool for easy functional annotation of proteomes and transcriptomes. Proteomics 17, 1700071. https://doi.org/10.1002/pmic.201700071.CrossRefGoogle ScholarPubMed
Castillo, JC, Shokal, U and Eleftherianos, I (2013) Immune gene transcription in Drosophila adult flies infected by entomopathogenic nematodes and their mutualistic bacteria. Journal of Insect Physiology 59, 179185. https://doi.org/10.1016/j.jinsphys.2012.08.003.CrossRefGoogle ScholarPubMed
Castillo, JC, Creasy, T, Kumari, P, Shetty, A, Shokal, U, Tallon, LJ and Eleftherianos, I (2015) Drosophila anti-nematode and antibacterial immune regulators revealed by RNA-Seq. BMC Genomics 16, 519. https://doi.org/10.1186/s12864-015-1690-2.CrossRefGoogle ScholarPubMed
Dziedziech, A, Shivankar, S and Theopold, U (2020) Drosophilamelanogaster responses against entomopathogenic nematodes: focus on hemolymph clots. Insects 11, 62. https://doi.org/10.3390/insects11010062.CrossRefGoogle Scholar
Eliáš, S, Hurychová, J, Toubarro, D, Frias, J, Kunc, M, Dobes, P, Simoes, N and Hyrsl, P (2020) Bioactive excreted/secreted products of entomopathogenic nematode heterorhabditis bacteriophora inhibit the phenoloxidase activity during the infection. Insects 11, 119. https://doi.org/10.3390/insects11060353.CrossRefGoogle ScholarPubMed
Garriga, A, Morton, A and Garcia-del-Pino, F (2018) Is Drosophila suzukii as susceptible to entomopathogenic nematodes as Drosophila melanogaster? Journal of Pesticide Science 91, 789798. https://doi.org/10.1007/s10340-017-0920-6.Google Scholar
Garriga, A, Mastore, M, Morton, A, Garcia-del-Pino, F and Brivio, MF (2020 a) Immune response of Drosophila suzukii larvae to infection with the nematobacterial complex Steinernema carpocapsaeXenorhabdus nematophila. Insects 11, 210. https://doi.org/10.3390/insects11040210.CrossRefGoogle ScholarPubMed
Garriga, A, Morton, A, Ribes, A and Garcia-del-Pino, F (2020 b) Soil emergence of Drosophila suzukii adults: a susceptible period for entomopathogenic nematodes infection. Journal of Pesticide Science 93, 639646. https://doi.org/10.1007/s10340-019-01182-w.Google Scholar
Garriga, A, Toubarro, D, Simões, N, Morton, A and Garcia-del-Pino, F (2023) The modulation effect of the Steinernema carpocapsaeXenorhabdus nematophila complex on immune-related genes in Drosophila suzukii larvae. Journal of Invertebrate Pathology 196, 107870. https://doi.org/10.1016/j.jip.2022.107870.CrossRefGoogle ScholarPubMed
Gress, BE and Zalom, FG (2019) Identification and risk assessment of spinosad resistance in a California population of Drosophila suzukii. Pest Management Science 75, 12701276. https://doi.org/10.1002/ps.5240.CrossRefGoogle Scholar
Huot, L, George, S, Girard, PA, Severac, D, Negre, N and Duvic, B (2019) Spodoptera frugiperda transcriptional response to infestation by Steinernema carpocapsae. Scientific Reports 9, 113. https://doi.org/10.1038/s41598-019-49410-8.CrossRefGoogle ScholarPubMed
Jones, K, Tafesh-Edwards, G, Kenney, E, Toubarro, D, Simoes, N and Eleftherianos, I (2022) Excreted secreted products from the parasitic nematode Steinernema carpocapsae manipulate the Drosophila melanogaster immune response. Scientific Reports 12, 113. https://doi.org/10.1038/s41598-022-18722-7.CrossRefGoogle ScholarPubMed
Kacsoh, BZ and Schlenke, TA (2012) High hemocyte load is associated with increased resistance against parasitoids in Drosophila suzukii, a relative of D. melanogaster. PLoS One 7, e34721. https://doi.org/10.1371/journal.pone.0034721.CrossRefGoogle ScholarPubMed
Kucerova, L, Broz, V, Arefin, B, Maaroufi, HO, Hurychova, J, Strand, H, Zurovec, M and Theopold, U (2016) The Drosophila Chitinase-like protein IDGF3 is involved in protection against nematodes and in wound healing. Journal of Innate Immunity 8, 199210. https://doi.org/10.1159/000442351.CrossRefGoogle ScholarPubMed
Lee, JC, Bruck, DJ, Curry, H, Edwards, D, Haviland, DR, Van Steenwyk, RA and Yorgey, BM (2011) The susceptibility of small fruits and cherries to the spotted-wing drosophila, Drosophila suzukii. Pest Management Science 67, 13581367. https://doi.org/10.1002/ps.2225.CrossRefGoogle Scholar
Lemaitre, B and Hoffmann, J (2007) The host defense of Drosophila melanogaster. Annual Review of Immunology 25, 697743. https://doi.org/10.1146/annurev.immunol.25.022106.141615.CrossRefGoogle ScholarPubMed
Li, B and Dewey, CN (2011) RSEM: accurate transcript quantification from RNA-Seq data with or without a reference. BMC Bioinformatics 12, 323. https://doi.org/10.1201/b16589.CrossRefGoogle ScholarPubMed
Li, E, Qin, J, Feng, H, Li, J, Li, X, Nyamwasa, I, Cao, Y, Ruan, W, Li, K and Yin, J (2021) Immune-related genes of the larval Holotrichia parallela in response to entomopathogenic nematodes Heterorhabditis beicherriana LF. BMC Genomics 22, 119. https://doi.org/10.1186/s12864-021-07506-4.Google ScholarPubMed
Lindgren, M, Riazi, R, Lesch, C, Wilhelmsson, C, Theopold, U and Dushay, MS (2008) Fondue and transglutaminase in the Drosophila larval clot. Journal of Insect Physiology 54, 586592.CrossRefGoogle ScholarPubMed
Liu, Y, Zhu, L, Guo, Z, Zhu, HD, Huang, ZH, Cao, HH, Yu, HZ, Liu, SH and Xu, JP (2022) Bombyx mori ferritin heavy-chain homolog facilitates BmNPV proliferation by inhibiting reactive oxygen species-mediated apoptosis. International Journal of Biological Macromolecules 217, 842852. https://doi.org/10.1016/j.ijbiomac.2022.07.169.CrossRefGoogle Scholar
Love, M, Huber, W and Anders, S (2014) Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biology 15, 550. https://doi.org/10.1186/s13059-014-0550-8.CrossRefGoogle ScholarPubMed
Mastore, M and Brivio, MF (2008) Cuticular surface lipids are responsible for disguise properties of an entomoparasite against host cellular responses. Developmental & Comparative Immunology 32, 10501062. https://doi.org/10.1016/j.dci.2008.02.003.CrossRefGoogle ScholarPubMed
McIntire, CR, Yeretssian, G and Saleh, M (2009) Inflammasomes in infection and inflammation. Apoptosis 14, 522535. https://doi.org/10.1007/s10495-009-0312-3.CrossRefGoogle ScholarPubMed
Mérel, V, Gibert, P, Buch, I, Rodriguez-Rada, V, Estoup, A, Gautier, M, Fablet, M, Boulesteix, M and Vieira, C (2021) The worldwide invasion of Drosophila suzukii is accompanied by a large increase of transposable element load and a small number of putatively adaptive insertions. Molecular Biology and Evolution 38, 42524267. https://doi.org/10.1093/molbev/msab155.CrossRefGoogle Scholar
Mishra, R, Chiu, J, Hua, G, Tawari, NR, Adang, MJ and Sial, AA (2018) High throughput sequencing reveals Drosophila suzukii responses to insecticides. Insect Science 25, 928945. https://doi.org/10.1111/1744-7917.12498.CrossRefGoogle ScholarPubMed
Moriya, Y, Itoh, M, Okuda, S, Yoshizawa, AC and Kanehisa, M (2007) KAAS: an automatic genome annotation and pathway reconstruction server. Nucleic Acids Research 35, 182185. https://doi.org/10.1093/nar/gkm321.CrossRefGoogle ScholarPubMed
Ogier, JC, Pagès, S, Frayssinet, M and Gaudriault, S (2020) Entomopathogenic nematode-associated microbiota: from monoxenic paradigm to pathobiome. Microbiome 8, 117. https://doi.org/10.1186/s40168-020-00800-5.CrossRefGoogle ScholarPubMed
Peña, JM, Carrillo, MA and Hallem, EA (2015) Variation in the susceptibility of Drosophila to different entomopathogenic nematodes. Infection and Immunity 83, 11301138. https://doi.org/10.1128/IAI.02740-14.CrossRefGoogle ScholarPubMed
R Core Team (2017) R: A Language and Environment for Statistical Computing. Vienna, Austria: R Found. Stat. Comput.Google Scholar
Schwanitz, T, Polashock, J, Stockton, D, Rodriguez-Saona, C, Sotomayor, D, Loeb, G and Hawkings, C (2022) Molecular and behavioral studies reveal differences in olfaction between winter and summer morphs of Drosophila suzukii. PeerJ 10, e13825. https://doi.org/10.7717/peerj.13825.CrossRefGoogle Scholar
Shan, T, Wang, Y, Bhattarai, K and Jiang, H (2023) An evolutionarily conserved serine protease network mediates melanization and toll activation in Drosophila. Science Advances 9, eadk2756. https://doi.org/10.1126/sciadv.adk2756.CrossRefGoogle ScholarPubMed
Shearer, PW, West, JD, Walton, VM, Brown, PH, Svetec, N and Chiu, JC (2016) Seasonal cues induce phenotypic plasticity of Drosophila suzukii to enhance winter survival. BMC Ecology 16, 118. https://doi.org/10.1186/s12898-016-0070-3.CrossRefGoogle Scholar
Sorensen, J, Nielsen, M, Kruhoffer, M, Justesen, J and Loeschcke, V (2005) Full genome gene expression analysis of the heat stress response in Drosophila melanogaster. Cell Stress & Chaperones 10, 312328. https://doi.org/10.1379/csc-128r1.1.CrossRefGoogle ScholarPubMed
Toubarro, D, Avila, MM, Montiel, R and Simões, N (2013) A pathogenic nematode targets recognition proteins to avoid insect defenses. PLoS One 8, 113. https://doi.org/10.1371/journal.pone.0075691.CrossRefGoogle ScholarPubMed
Walsh, DB, Bolda, MP, Goodhue, RE, Dreves, AJ, Lee, J, Bruck, DJ, Walton, VM, O'Neal, SD and Zalom, FG (2011) Drosophila suzukii (Diptera: Drosophilidae): invasive pest of ripening soft fruit expanding its geographic range and damage potential. Journal of Integrated Pest Management 2, 17. https://doi.org/10.1603/IPM10010.CrossRefGoogle Scholar
Wang, X, Daane, KM, Hoelmer, KA and Lee, JC (2021) Biological control of spotted-wing Drosophila: an update on promising agents. In Mello Garcia, FR (ed.), Drosophila suzukii Management. Switzerland: Springer Nature, pp. 143168.Google Scholar
Woodring, JL and Kaya, HK (1988) Steinernematid and Heterorhabditid Nematodes: A Handbook of Biology and Techniques. Southern Cooperative Series Bulletin 331. Fayetteville, AR: Arkansas Agricultural Experiment Station.Google Scholar
Wu, T, Hu, E, Xu, S, Chen, S, Chen, M, Guo, P, Dai, Z, Feng, T, Zhou, L, Tang, W, Zhan, L, Fu, X, Liu, S, Bo, X and Yu, G (2021) Clusterprofiler 4.0: a universal enrichment tool for interpreting omics data. Innovation 2, 100141. https://doi.org/10.1016/j.xinn.2021.100141.Google ScholarPubMed
Xing, S, Deng, D, Wen, W and Peng, W (2022) Functional transcriptome analyses of Drosophila suzukii midgut reveal mating-dependent reproductive plasticity in females. BMC Genomics 23, 726. https://doi.org/10.1186/s12864-022-08962-2.CrossRefGoogle Scholar
Yadav, S, Daugherty, S, Shetty, AC and Eleftherianos, I (2017) RNAseq analysis of the Drosophila response to the Entomopathogenic Nematode Steinernema. Genes|Genomes|Genetics 7, 19551967. https://doi.org/10.1534/g3.117.041004.CrossRefGoogle Scholar
Yadav, S, Frazer, J, Banga, A, Pruitt, K, Harsh, S, Jaenike, J and Eleftherianos, I (2018) Endosymbiont-based immunity in Drosophila melanogaster against parasitic nematode infection. PLoS One 13, 120. https://doi.org/10.1371/journal.pone.0192183.CrossRefGoogle ScholarPubMed
Figure 0

Table 1. Summary statistics of transcriptome sequencing

Figure 1

Figure 1. Volcano plot representation of the differentially expressed genes comparing the transcriptome of infected and control larvae.

Figure 2

Figure 2. Significantly enriched terms of Gene Ontology (GO) after the analysis of the DEG using TopGO, considering the categories: biological process (BP), Molecular Function (MF), and Cell Component (CC).

Figure 3

Figure 3. Significantly enriched terms (x-axis) using the KEGG database and the transcript count (y-axis) after the analysis of the DEG, considering (A) the level 2 pathways and (B) the level 3 genes.

Figure 4

Table 2. List of significant DEG associated with immune defence responses upon S. carpocapsae infection, with the mRNA identification (ID RNA), the protein identification (ID Protein), and the gene name obtained during the annotation step

Supplementary material: File

Garriga et al. supplementary material 1

Garriga et al. supplementary material
Download Garriga et al. supplementary material 1(File)
File 921.8 KB
Supplementary material: File

Garriga et al. supplementary material 2

Garriga et al. supplementary material
Download Garriga et al. supplementary material 2(File)
File 61.1 KB