Abstract
Drosophila immunity has been the focus of intense study and has impacted other research fields including innate immunity and agriculturally or epidemiologically relevant investigations of insect pests and vectors. Unsurprisingly for such a large body of work, some published results were later found to be irreproducible. Although some results have been contradicted in the literature, many have no published follow-up, either due to a lack of research or low motivation to publish negative or contradictory results. We have addressed this by performing a reproducibility project that analyses the verifiability of claims from articles published on Drosophila immunity before 2011. To assess reproducibility, we extracted claims from 400 articles on the Drosophila immune response to bacteria and fungi and performed preliminary verification by comparing these claims to other published literature in the field. Using alternative approaches, we also experimentally tested some ‘unchallenged’ claims, which had no published follow-up. The intent of this analysis was to centralize evidence on insights and findings to improve clarity for scientists that may base research programs on these data. All our data are published on a publicly available website associated with this article (https://ReproSci.epfl.ch/) that encourages community participation. This article provides a short summary of claims that were found to have contradictory evidence, which may help the community to assess past findings on Drosophila immunity and improve clarity going forward.
Introduction
Innate immune mechanisms have become central to our understanding of immunology (Danilova, 2006; Medzhitov and Iwasaki, 2024; Pradeu et al., 2024), although their study was long overshadowed by interest in adaptive immunity. Innate immune mechanisms are now widely recognized as having key importance as a first layer of defense, in the recognition of infectious agents, and in subsequent polarization of the immune response. These responses are activated by a specific set of receptors that sense microbe-specific molecules, virulence factors and/or infection-induced damage. Complex signaling pathways integrate this information to induce specific innate immune modules adapted to the characteristics of the encountered pathogen. Characterization of the innate immune response has greatly benefitted from the use of model organisms such as Drosophila, which is amenable to genetic studies and relies exclusively on innate immunity for defense (Buchmann, 2014; Westlake et al., 2024).
The Drosophila immune system is composed of several immune modules, some of which are conserved in vertebrates (Buchon et al., 2014; Liegeois and Ferrandon, 2022; Westlake et al., 2024). The systemic immune response is best characterized by the production and secretion into the hemolymph of host-defense peptides by the fat body and hemocytes. The best-characterized of these effectors are the antimicrobial peptides (AMPs). AMPs are small cationic peptides with antimicrobial activity, whose expression is induced to incredibly high levels in response to infection (Hanson and Lemaitre, 2020). The systemic antimicrobial response is predominantly regulated at the transcriptional level by the Toll and Imd signaling pathways. In Drosophila, the Toll pathway is activated by microbial cell wall components (fungal glucans and Lysine-type peptidoglycan), and microbial proteases. Toll signaling culminates in the activation of two NF-κB transcription factors, Dif (dorsal-related immunity factor) and Dorsal, which regulate a large set of immune genes such as the Bomanins, the antifungal peptide Drosomycin, and many other proteins (Clemmons et al., 2015; De Gregorio et al., 2002; Troha et al., 2018). Flies lacking Toll pathway activity are viable but highly susceptible to infection by Gram-positive bacteria and fungi (Lemaitre et al., 1996). The Imd pathway is activated by DAP-type peptidoglycans produced by Gram-negative bacteria and a subset of Gram-positive bacteria (e.g. Bacillus) (Kaneko et al., 2004; Leulier et al., 2003). Binding of peptidoglycan to receptors of the Peptidoglycan Recognition Protein family (PGRP-LC, PGRP-LE, PGRP-SD) initiates an intracellular signaling cascade with homology to the TNFα-Receptor pathway, which ultimately activates the NF-κB factor Relish (Choe et al., 2002; Gottar et al., 2002; Iatsenko et al., 2016; Takehana et al., 2004). The Imd pathway regulates the expression of many genes encoding antibacterial peptides, serine proteases, and the iron sequestration protein Transferrin, among others. Imd deficient flies are viable, but susceptible to Gram-negative bacterial infection.
Complementary to these transcriptional programs, melanization is an arthropod-specific immune response resulting in rapid deposition of the black pigment melanin at wound or infection sites and concomitant production of microbicidal reactive oxygen species (Cerenius et al., 2008). Melanization relies on the activation of enzymes called prophenoloxidases (PPOs), which catalyze the oxidation of phenols, resulting in melanin polymerization (Dudzic et al., 2015). This elegant process also hardens clots with melanin polymer plugs that prevent blood loss, akin to the mechanical function of fibrin scabs. Clotting, which involves both humoral factors and hemocytes, contributes to wound healing and has been implicated in host defense against nematodes (Theopold et al., 2014; Yadav and Eleftherianos, 2018).
In addition to soluble immune effectors, cellular components also play a crucial role in immunity. In adult flies, two hemocyte types known as plasmatocytes and crystal cells are present in circulation or as sessile cells attached to the inner cuticle (Banerjee et al., 2019; Lanot et al., 2000). Plasmatocytes are macrophage-like cells with phagocytic activity, while crystal cells produce and store PPO enzymes and contribute to melanization. Use of hemocyte deficient flies has shown that hemocytes contribute to survival upon systemic infection to certain bacterial species, and several phagocytic receptors have been identified (Charroux and Royet, 2009; Defaye et al., 2009; Melcarne et al., 2019a; Stephenson et al., 2022; Ulvila et al., 2011).
Natural infection with microbes also triggers a local immune response in epithelia, which has been extensively characterized in the gut (Buchon et al., 2013; Tafesh-Edwards and Eleftherianos, 2023; Westlake et al., 2024). In this organ, a range of defenses including the peritrophic matrix, an acid pH, peristalsis, and epithelial repair through stem cell proliferation all contribute to deterring ingested pathogens. Local production of AMPs and ROS provide two complementary inducible defense mechanisms in the gut (Liehl et al., 2006; Ryu et al., 2006). It is now clear that each organ contributes individual input to the immune response in an integrated manner, which vary according to the route of infection and the nature of the pathogens (Westlake et al., 2024). Finally, extensive studies have highlighted the role of the immune system upon mating and in the reproductive organs to prevent infection (Schwenke et al., 2016; Siva-Jothy, 2009).
Drosophila immunity has been a topic of intense research, with thousands articles and reviews published on this topic (Westlake et al., 2024). Characterization of the Drosophila immune system has contributed significantly to fields including innate immunity and agriculturally or epidemiologically relevant investigations of insect pests and vectors. As expected in such a large body of work, some published results were later found to be irreproducible. Although some of these results have been contradicted in the literature, many have no published follow-up, either due to a lack of research or low motivation to publish negative or contradictory results. We have addressed this by performing an extensive reproducibility project that analyses the validity of claims from articles on Drosophila immunity accepted for publication before 2011. This time frame allows opportunity for publication of follow-up results. We extracted major and minor claims from 400 articles on the Drosophila immune response to bacteria and fungi and performed verification where possible by comparing to other published literature in the field. We also experimentally tested 55 ‘unchallenged’ claims with no published follow-up, a process that will be continuously expanded with the help of the community.
The intent of this analysis is not to assign merit to individual scientists or publications, but rather to centralize evidence on results or groups of results to improve clarity for scientists that may base their research on these data. Although conflicting data are often discussed at conferences or at less formal meetings, diffusion of this knowledge is limited due to lack of publications. Our hope is to provide a dataset with community-sourced proofs that allow a wider range of scientists to easily identify claims with contradictory evidence. In association with this article, we have published the ReproSci website, which describes and presents assessments of claims from 400 articles and encourages community participation (https://ReproSci.epfl.ch/). This article synthesizes the main results on claims of particular interest discovered during the reproducibility analysis, while the website provides a more complete analysis. These include claims published in high profile journals that have been contradicted or have tenuous support. This article is accompanied by 28 supplementary materials that include new experimental data from various laboratories that support or contradict some unresolved claims that are discussed in this article. In many cases, re-analysis of these claims is made possible by advances in understanding of immune mechanisms and technology since publication of the initial articles. Statistical analysis of this reproducibility study and metascience insights obtained from this approach are discussed in a companion article.
Results
Overview of the reproducibility project
400 research articles focusing on Drosophila immunity published between 1959 and 2011 were selected by PubMed keyword retrieval and manual curation (Supplementary Table 1). Articles dealing with population genetics, virulence of entomopathogens, and the immune response to viruses and wasps were excluded. The main (1 per article), major (2-4 per article) and minor (0-4 per article) claims were extracted along with a summary of evidence supporting the claims and the methods of each article. The reproducibility of each claim was then assessed by analyzing subsequent publications on the topic from the same or other laboratories. Claims were assessed as verified, unchallenged, challenged, mixed, or partially verified (see Methods). All article summaries and assessments were released to the community on July 11th, 2023, via the ReproSci website, which allows users to comment on the claims and provide confirmatory or contradictory evidence. 45 major and 11 minor claims (many of which are discussed in the Supplement) were also experimentally tested in various laboratories using alternate approaches. For instance, a result initially obtained using RNAi may be tested using newly generated CRISPR mutants. Our project focuses on assessing the strength of the claims themselves (inferential/indirect reproducibility) rather than testing whether the original methods produce repeatable results (results/direct reproducibility). Thus, our conclusions do not directly challenge the initial results leading to a claim, but rather the general applicability of the claim itself. This article presents some of the main observations from the ReproSci project and is intended to allow the reader to quickly grasp key claims in the Drosophila immunity field with low reproducibility (challenged claims) or not as simple as proposed in the initial articles.
Disclaimer
The aim of the ReproSci project is to assess the replicability of claims made in articles published in the field of Drosophila immunity. This is in no way an assessment of the ‘scientific value’ of the research. Although a lack of replicability is often viewed negatively, papers that include non-replicable observations may nevertheless have a positive impact by developing new techniques, introducing interesting concepts, providing a foundation for future discoveries, or presenting results that are useful despite incorrect interpretation. Similarly, articles that include only well-supported claims may be of low scientific value if they only confirm previous observations and present little in the way of innovation. The leading authors (H.W., B.L.) acknowledge that the selection of claims within papers and assessments of those claims cannot be fully objective. Opening the ReproSci database to comment by community members is intended to provide a greater degree of objectivity and consensus and generate ongoing discussion beyond initial assessments.
Insights from the ReproSci project on the Toll pathway
Toll is not regulated by 18-wheeler
Following the characterization of the Toll receptor (Toll-1) as a key regulator of Drosophila AMP genes (Lemaitre et al., 1996), the Toll homolog 18-wheeler (Toll-2) was reported to have a complementary role in LPS sensing and regulation of the antibacterial response, similar to mammalian TLR4 (Williams et al., 1997). This paper was initially widely cited, but was directly challenged by a follow-up study revealing that 18-wheeler does not have a direct role in immunity, but rather indirectly affects survival and AMP gene expression through a general negative impact on fly development (Ligoxygakis et al., 2002).
GNBP1 is not a sensor of fungal glucan
GNBP1 (Gram-Negative Binding Protein 1) was initially characterized for its ability to bind LPS and fungal glucan, based on overexpression in cell culture and in vitro binding studies (Kim et al., 2000). Subsequent genetic studies revealed that this secreted protein instead cooperates with PGRP-SA in sensing of Gram-positive bacteria, and find no evidence that GNBP1 acts as an immune sensor for LPS or glucan (Gobert et al., 2003; Pili-Floury et al., 2004). GNBP3, a Drosophila homolog of GNBP1, does have glucan binding activity and activates immune responses to fungi in vivo (Gottar et al., 2006).
GNBP1 does not process peptidoglycan
Gram-positive bacteria stimulate the Toll pathway through cooperative action of PGRP-SA and GNBP1, which activate the apical serine protease ModSP upon sensing of peptidoglycans (Buchon et al., 2009b; Gobert et al., 2003; Pili-Floury et al., 2004). Structural studies revealed that PGRP-SA can bind peptidoglycan, consistent with its demonstrated role as pattern-recognition receptor in vivo (Chang et al., 2004). However, one study suggested that GNBP1 can also digest peptidoglycan, and that this hydrolytic function of GNBP1 is required for full activation of PGRP-SA and ModSP (Wang et al., 2006). Reported enzymatic activity of GNBP1 has never received follow up. Studies in other insects such as Tenebrio molitor and Manduca sexta suggest that the primary function of GNBP1 is as an adaptor between PGRP-SA and the apical serine protease ModSP (Kim et al., 2008; Park et al., 2007; Wang et al., 2022). In vitro experiments performed as part of ReproSci found that GNBP1 does not process peptidoglycan in the absence or presence of PGRP-SA (Supplementary S1). Similarly, pre-incubation of GNBP1 with peptidoglycan did not enhance ModSP autoactivation. These results indicate that contrary to the initial claim, GNBP1 has no enzymatic activity against peptidoglycan. The initial results may have been obtained due to residual impurities in preparations of recombinant GNBP1.
Spirit, Sphinx and Spheroide are not key components of the Toll pathway
An RNAi screen identified five serine proteases involved in the activation of the Toll ligand Spatzle during the immune response (Kambris et al., 2006a). The roles of two of these genes, SPE and grass, were confirmed in subsequent studies, although grass was shown to affect the whole pattern recognition branch of the Toll pathway and not only the PGRP-SA/GNBP1 branch as suggested in the initial study (El Chamy et al., 2008; Jang et al., 2006). New data from ReproSci using viable loss-of-function mutations and recent-generation RNAi reveals that of the remaining three genes, neither the serine protease homologs Spheroide and Sphinx1/2 nor the serine protease Spirit are strictly required for Toll activation (Supplementaries S2, S3, S4). It is likely that Sphinx1/2 has no role in the immune response, while the immune-inducible gene Spirit may have a redundant or a subtle role. An independent study suggests Spheroide has a subtle role in Toll activation (Patrnogic and Leclerc, 2017); experiments performed for ReproSci support these results. Non-replicable results on the roles of Spirit, Sphinx and Spheroide in Toll pathway activation may be due to off-target effects common to first-generation RNAi tools (Ueda, 2001).
The Toll immune pathway is not negatively regulated by wntD
Gordon et al., (2005a) found that wntD acts as a feedback inhibitor of dorsal but not dif during the innate immune response. wntD mutants had elevated expression of Diptericin but not Drosomycin in response to septic infection with the Gram-positive bacterium Micrococcus luteus, and increased susceptibility to Listeria monocytogenes that was rescued in wntD, dorsal double mutants. However, data from FlyAtlas (Thurmond et al., 2019) show that wntD is only expressed at the embryonic stage and not at the adult stage at which the experiments were performed by (Gordon et al., 2005a). Using an independent null mutant of wntD, we did not observe any impact of wntD on expression of Diptericin and Drosomycin, or increased sensitivity to Listeria monocytogenes infection (Supplementary S5). Thus, we conclude that wntD is not a negative regulator of the Toll pathway during the immune response. The initial claim concerning wntD may be explained by a genetic background effect independent of wntD.
Toll is not activated by overexpression of GNBP3 or Grass
Experiments performed for ReproSci find that contrary to previous reports, overexpression of GNBP3 (Gottar et al., 2006) or Grass (El Chamy et al., 2008) in the absence of immune challenge does not effectively activate Toll signaling (Supplementaries S6, S7).
Insights from the reproducibility project on the Imd pathway
PGRP-SD is a component of Imd and not Toll signalling
PGRP-SD was initially identified as a secreted pattern-recognition receptor involved in the Toll pathway in response to certain Gram-positive bacteria (Bischoff et al., 2004; Wang et al., 2008). This was inconsistent with subsequent observations that i) PGRP-SD binds DAP-type peptidoglycan found in Gram-negative bacteria which typically activate Imd signaling (Basbous et al., 2011; Leone et al., 2008; Lim et al., 2006); and that ii) this gene is regulated by the Imd pathway (De Gregorio et al., 2002). Using isogenic fly stocks and newly generated mutants, Iatsenko et al., subsequently showed that PGRP-SD is a positive regulator of the Imd pathway upstream of PGRP-LC that promotes sensing of DAP-peptidoglycan (Iatsenko et al., 2016). Analysis of several fly stocks expected to carry the PGRP-SD∆S3mutation used in the initial study revealed the presence of a wild-type copy PGRP-SD, suggesting that either the stock used in this study did not carry the expected mutation, or that the mutation was lost by contamination prior to sharing the stock with other labs. Additional in vitro experiments performed in the framework of ReproSci reveal that PGRP-SD does not contribute to Toll activation by peptidoglycan under experimental conditions (Supplementary S1). This suggests that the original results assigning PGRP-SD to the Toll pathway may be due to a genetic background effect.
PGRP-LE is exclusively intracellular and does not participate in melanization
(Takehana et al., 2004, 2002) found that intracellular PGRP-LE contributes to the activation of the Imd pathway together with the transmembrane receptor PGRP-LC. While its role as an intracellular sensor has been confirmed (Bosco-Drayon et al., 2012; Neyen et al., 2012), the initial papers claimed that an extracellular form of PGRP-LE could function akin to CD14 (Takehana et al., 2004, 2002). Follow-up papers have not addressed the existence of this extracellular form. Using a new proteomic approach, we did not detect PGRP-LE in adult fly hemolymph (Rommelaere et al., 2024). Moreover, use of a GFP tagged version of PGRP-LE (Bosco-Drayon et al., 2012) did not reveal an extracellular form in larvae (Supplementary S8). We conclude that PGRP-LE is an exclusively intracellular sensor, with a prominent role regulating the gut immune response (Bosco-Drayon et al., 2012; Neyen et al., 2012). In addition, experiments performed for ReproSci found no role for PGRP-LE in melanization, contrary to (Takehana et al., 2004, 2002) (Supplement S9). The results of the initial papers are likely due to the use of ubiquitous overexpression of PGRP-LE, resulting in melanization due to overactivation of the Imd pathway and resulting tissue damage. We were also unable to find a role for PGRP-LE in survival to Bacillus infections (Supplementary S10) and conclude that the initial result was due to genetic background and comparison to a robust wild-type as the baseline.
LPS does not activate the Imd pathway
In mammals, LPS is a strong inducer of TLR4-NF-κB pathway (Hoshino et al., 1999). Studies in Drosophila using LPS from SigmaTM initially provided evidence that LPS can stimulate the Imd pathway (Imler et al., 2000). However, use of highly purified bacterial cell wall components showed that DAP-type peptidoglycan and not LPS stimulates Imd pathway activity (Kaneko et al., 2004; Leulier et al., 2003). Furthermore, in contrast to an initial claim by Leulier et al., 2003, monomeric DAP-type peptidoglycan (notably tracheal cytotoxin, TCT) is also a strong elicitor of the Imd pathway, in addition to polymeric peptidoglycan (Kaneko et al., 2004; Stenbak et al., 2004).
Naked DNA is not immunogenic in Drosophila
In mammals, free DNA can activate IFN-mediated inflammatory responses through sensors such as cGAS (Decout et al., 2021). In 2002, an article reported that DNA is immunogenic in Drosophila after observing elevated expression of Diptericin genes in DNase IIlo hypomorphic mutant flies (Mukae et al., 2002). However, experiments performed for ReproSci using the original DNAse IIlo hypomorph show that elevated Diptericin expression in the hypomorph is eliminated by outcrossing of chromosome II, and does not occur in an independent DNAse II null mutant, indicating that this effect is due to genetic background (Supplementary S11). Furthermore, elevated Diptericin expression in DNAse IIlo flies is not observed in germ-free conditions, indicating that it is not due to DNA excess (West, 2018). Moreover, experiments showed that injection of naked bacterial and eukaryotic DNA does not activate an immune response in wild-type or DNAse II mutant flies. These results indicate that naked DNA unlikely to be an elicitor for AMP genes in Drosophila as originally claimed, although it may activate the recently discovered cGLR-STING pathway (Cai et al., 2022). The possibility that DNA-associated proteins such as histones are immunogenic also remains to be investigated.
JNK signaling is not directly required for AMP expression
Several studies pointed to a direct role of the JNK pathway in the regulation of AMPs (Delaney et al., 2006; Kallio et al., 2005; Kleino et al., 2005) while other studies did not identify any major role (Boutros et al., 2002; Kenmoku et al., 2017; Park et al., 2004; Silverman et al., 2003). Experiments performed for ReproSci and review of the literature suggest that JNK is unlikely to have a strong direct or systematic role on regulation of Drosophila AMPs in vivo, and is not required in the adult fat body for AMP expression in response to infection (Supplementary S12). Previous results finding a direct role for JNK in AMP production are likely due to a combination of i) use of cell culture systems that do not fully reflect in vivo systems; ii) the incorrect conclusion that kinase-dead JNK kinase (TAK1) is biologically equivalent to a null mutant; and iii) underappreciation for the overarching role that JNK plays in cytoskeletal remodeling, pathway activation, and cell differentiation, which are not specific to immunity.
Caspar is not a canonical negative regulator of Imd signaling
caspar encodes an orthologue of human Fas associated factor 1 (FAF1) which was reported to interact with key immune proteins including NF-κB. Kim et al., (2006) found that a null mutation of caspar produced high constitutive expression of Diptericin and increased resistance to Gram-negative infections. Using flies deficient for caspar, we did not detect any increased Diptericin expression in unchallenged flies as initially reported (Supplementary S13). Moreover, Caspar overexpression did not result in significantly lower levels of Diptericin, and did not promote survival to Gram-negative infections. We conclude that while Caspar might affect the Imd pathway in certain tissue-specific contexts, it is unlikely to act as a generic negative regulator of the Imd pathway.
Insights from the reproducibility project on immune-induced peptides
Edin does not significantly contribute to lifespan or resistance against L. monocytogenes
Gordon et al., (2008) identified edin, a host defense peptide gene highly expressed upon L. monocytogenes infection. They found that overexpression of edin markedly reduced lifespan and fly fitness whereas RNAi knockdown of edin in the fat body increased susceptibility to L. monocytogenes infection. Contrary to (Gordon et al., 2008), experiments performed for ReproSci using a novel edin null mutant indicate that loss of edin does not increase susceptibility to L. monocytogenes infection compared to appropriate controls. Moreover, overexpression of edin did not cause any major reduction in lifespan (Supplementary S14).
Listericin overexpression is not protective against L. monocytogenes infection
(Goto et al., 2010) found using transfection and RNAi in S2 cells that the Listericin gene encodes a novel antibacterial peptide-like protein whose induction is cooperatively regulated by PGRP-LE and the JAK-STAT pathway. However, a microarray indicates that this gene is regulated by the Imd pathway with a minor impact of Toll (De Gregorio et al., 2002). Experiments performed for ReproSci suggest that Listericin expression does not specifically depend on PGRP-LE but rather is induced through both PGRP-LC and -LE as other Imd-regulated AMPs are (Supplementary S15). Silencing STAT92A in the fat body by using an in vivo RNAi approach did not confirm a role of JAK/STAT in induction of Listericin following infection as claimed by (Goto et al., 2010). Overexpression of Listericin using a newly generated construct did not reveal any protective effect against L. monocytogenes as found by (Goto et al., 2010). Thus, Listericin should be considered as a host defense peptide regulated by the Imd pathway whose activity remains to be confirmed.
Insights from the reproducibility project on phagocytosis and encapsulation
Dscam1 does not directly contribute to phagocytosis through opsonization
Drosophila Dscam1 has three immunoglobulin-like domains and encodes thousands of isoforms through alternate splicing. An early paper proposed that secreted Dscam1 isoforms can bind bacteria and enhance phagocytosis, an attractive hypothesis due to its potential to produce multiple ‘recognition’ isoforms akin to an adaptive immune system (Watson et al., 2005). Dscam1 homologs have independently been implicated in crustacean and mosquito immunity, systems that are not amenable to genetic studies (Dong et al., 2006; Ng et al., 2014; Ng and Kurtz, 2020). However, no follow-up studies were published on the role of Dscam1 proteins in Drosophila immunity. Subsequent studies showed that Dscam1 splicing is not affected by septic injury (Armitage et al., 2014), and contrary to a role for secreted Dscam1 isoforms in Drosophila opsonization, a proteomic analysis of adult hemolymph did not detect any secreted Dscam1 isoforms (Rommelaere et al., 2024). Furthermore, all Dscam1 isoforms encode the transmembrane domain. Experiments performed for ReproSci did reveal a mild decrease in phagocytosis of E. coli and S. aureus by hemocytes of hml>Dscam1-RNAi larvae: a 30% reduction in comparison to 50% in the initial study (Watson et al., 2005) (Supplementary S16). Dscam1 plays a key role in neuronal dendrite arborization and affects how neurons influence peripheral hematopoiesis, and RNAi against Dscam1 in neurons reduces hemocyte numbers (Ouyang et al., 2020). It is likely that reduced phagocytosis in Dscam1 depleted animals is due to an effect on hematopoiesis rather than a direct immune effect. In the absence of additional studies, Dscam1 should not be considered a Drosophila immune gene sensu stricto.
PGRP-LC is not a phagocytic receptor
(Rämet et al., 2002) reported a role of the Imd receptor PGRP-LC in phagocytosis of Gram-negative bacteria in an extensive RNAi screen (Rämet et al., 2002). Subsequent studies did not reveal any critical role of PGRP-LC in bacterial engulfment (Choe et al., 2002; Melcarne et al., 2019a). It is likely that PGRP-LC indirectly (and modestly) affects phagocytosis through its role in the Imd pathway and associated upregulation of phagocytic receptors (Wong et al., 2017), but this pattern recognition receptor should not be considered a phagocytic receptor.
Croquemort is not a phagocytic engulfment receptor for apoptotic cells or bacteria
The CD36 scavenger member Croquemort was initially identified as a macrophage-specific phagocytic receptor for apoptotic cells. Use of an overlapping set of deficiencies removing croquemort revealed that Croquemort was critical for engulfment of apoptotic cells during embryogenesis (Franc, 1999; Franc et al., 1996). However, using a clean croquemort null mutant, experiments performed for ReproSci reveal that croquemort is not required for removal of apoptotic cells in embryos (Supplementary S17). croquemort mutant phagocytes do display a general defect in phagosome maturation and accumulate acidic vacuoles (similar to draper mutants) (Guillou et al., 2016; Han et al., 2014). It is not known why croquemort affects phagosome maturation, but could be due to its role in lipid uptake (Woodcock et al., 2015). The available data indicate that Croquemort is not a phagocytic receptor required for initial uptake but contributes to sustaining phagocytosis by promoting lysosomal degradation. Of note, another CD36 member, Santa Maria, was recently implicated in the phagocytosis of apoptotic cells by glia (Hilu-Dadia et al., 2025).
dSR-CI is not required for phagocytosis of bacteria
The scavenger receptor dSR-CI was initially identified as a hemocyte-restricted receptor with high affinity for low density lipoprotein (Pearson et al., 1995). In vitro and RNAi experiments suggested a significant role for dSR-CI in phagocytosis of bacteria (Rämet et al., 2001). Experiments for ReproSci using a newly generated dSR-CI null mutant did not reveal any major requirement of dSR-CI in phagocytosis in vivo (Supplementary S18). It is possible that dSR-CI has a redundant role in phagocytosis with one or more of the three additional dSR-Cs encoded in the Drosophila genome (Ulvila et al., 2011), or may play another role in hemocytes which remained to be determined. We confirm the (Rämet et al., 2001) observation that loss of SR-CI has no apparent effect on the inducible AMP response.
Retinophilin/Undertaker has no role in phagocytosis
Using overlapping deficiencies in embryos and RNAi in S2 cells, (Cuttell et al., 2008a) found that Undertaker (uta/rtp), a Junctophilin, was required for phagocytosis of apoptotic cells and bacteria and links Draper-mediated phagocytosis to Ca2+ homeostasis. However, expression of Undertaker is restricted to the adult eye (Thurmond et al., 2019), which argues against a role in phagocytosis by embryonic macrophages. Using an rtp1 null allele, we did not observe any effect of Undertaker on phagocytosis of apoptotic cells or bacteria by larval hemocytes (Supplementary S19). The published data are most consistent with a scenario in which RNAi generated off-target knockdown of a protein related to retinophilin/undertaker, while Undertaker itself is unlikely to have a role in phagocytosis. In contrast, independent data support the notion put forth by (Cuttell et al., 2008a) that calcium flux and store-operated calcium entry (including all other components mentioned in this paper) are required for phagocytosis and blood cell activation in animals (Etchegaray et al., 2012; Gronski et al., 2009; Moon et al., 2020; Mortimer et al., 2013; Weavers et al., 2016).
NimC1 has a co-operative role in phagocytosis of Gram-negative bacteria
An in vivo RNAi approach suggested that NimC1 is required for the uptake of Gram-negative bacteria (Kurucz et al., 2007). In contrast, a null mutation in NimC1 did not reproduce this phenotype (Melcarne et al., 2019). However a NimC1, eater double mutant revealed that these two Nimrod receptors function co-operatively in phagocytosis of Gram-negative bacteria (Melcarne et al., 2019b). It is possible this disparity relies on a cryptic effect, such as genetic background differences. Although the initial claim that NimC1 alone is essential for phagocytosis of Gram-negative bacteria was not reproducible, the claim that NimC1 is involved in phagocytosis of Gram-negative bacteria is correct.
Eater has a co-operative role in phagocytosis
An initial study pointed to a role for the transmembrane receptor Eater in the phagocytosis of both Gram-negative and Gram-positive bacteria (Kocks et al., 2005). The authors used an overlapping set of deficiencies to remove Eater in vivo and RNAi in S2 cells to reach this conclusion. While nearly all of the claims in the original article have been confirmed, subsequent studies showed that a null mutation of eater does not impact phagocytosis of Gram-negative bacteria (Bretscher et al., 2015; Melcarne et al., 2019). Reduced phagocytosis of Gram-negative bacteria was observed only in NimC1, eater double mutants, indicating that Eater is involved in this process but not strictly necessary.
Psidin is not required for induction of Defensin
Brennan et al., (2007) found that Psidin acts non-autonomously in the hemocytes to control induction of Defensin in the fat body, and suggested that a cytokine produced by hemocytes is required to activate systemic Defensin expression. In contrast to this, experiments performed for ReproSci using two independent psidin mutants show that Defensin expression is similar to wild type in these lines (Supplementary S20). Furthermore, evidence from multiple papers suggests that this result, and other instances where mutations have been found to specifically eliminate Defensin expression, is likely due to segregating polymorphisms within Defensin that disrupt primer binding in some genetic backgrounds and lead to a false negative result (Supplementary S20). (Brennan et al., 2007) further found that Psidin is required for phagolysosomal maturation. Experiments performed for ReproSci confirm that phagolysosomal maturation is impaired in independent psidin mutants, and furthermore that psidin mutant hemocytes are abnormally large and multinucleate (Supplementary S20).
Hemese is not essential for encapsulation
(Kurucz et al., 2003) found that loss of Hemese, a hemocyte-specific transmembrane protein, causes overproduction of lamellocytes in permissive conditions. In contrast to this, using an independent hemeseSK2null mutant, we find that loss of hemese does not strongly affect lamellocyte number or encapsulation (Supplementary S21). The function of Hemese awaits further functional characterization.
Insights from the reproducibility project on IRC, Duox and NOS
Immune Regulated Catalase (IRC) is not essential for survival to benign enteric infection
Using a ubiquitous and directed RNAi approach, (Ha et al., 2005b) found that adult flies depleted of the extracellular catalase IRC (immune regulated catalase) displayed high mortality rates after ingestion of foods contaminated with normally benign microbes (Ha et al., 2005b). Contrary to this, we did not observe any increase in mortality upon feeding IRC null mutant flies with various microbial pathogens, although an effect of the mutation on motor function and overall fitness was observed (Supplementary S22). The immune regulated gene IRC is likely to play a role in the immune response that remains to be characterized.
Duox-mediated ROS production is most likely a signalling mechanism rather than an antibacterial effector
Early experiments showed that ingestion of pathogenic bacteria by adult flies resulted in rapid upregulation of Duox expression in the midgut and an associated Duox-dependent reactive oxygen species (ROS) burst as shown by ubiquitous knockdown of this gene. Ha et al., (2005b) inferred that ROS produced by Duox, particularly HOCl, had highly bactericidal effects, as clearance of ingested bacteria was impaired in Duox-RNAi flies (Ha et al., 2005a). A role of Duox in the control of the gut microbiota has also been subsequently observed in other insects (Yao et al., 2016). Epithelium damage associated with Duox-mediated production of ROS was also thought to explain the increased epithelium renewal observed upon oral bacterial infection (Buchon et al., 2009a). However, most of these experiments were performed using a ubiquitous Gal4 driver to silence Duox. Duox is weakly expressed in the midgut compared to its expression in the adult crop, or larval hindgut and trachea (Thurmond et al., 2019), and is an essential gene required for development (Hurd et al., 2015). Experiments done in the framework of ReproSci were unable to reproduce a defect in bacterial clearance (Ecc15-GFP) in Duox-RNAi flies at the same time point as the initial publication (Supplementary S23). Furthermore, we did not observe any increase in Duox expression in the gut upon oral bacterial infection. Recent studies show that rather than Duox, the alternate Drosophila NADPH oxidase Nox stimulates stem cell proliferation upon bacterial infection (Iatsenko et al., 2018; Jones et al., 2013; Patel et al., 2019). Additionally, a recent article suggests that the impact of Duox on gut epithelial renewal is an indirect effect of Duox in the Malpighian tubules rather than the midgut (Liu et al., 2024). Other evidence suggests that lumenal ROS produced by Duox contributes to an early bacterial clearance via a signaling role that promotes visceral muscle contraction, regardless of its potential bacteriostatic activity (Benguettat et al., 2018; Du et al., 2016). We conclude that data on a bactericidal role for Duox in the gut context remain contradictory. More precise experiments are needed to clearly define the role of Duox in gut epithelial immunity, which may primarily play a signaling role rather than a microbicidal one.
Nitric oxide synthase (NOS) is not essential for systemic AMP expression upon oral infection
A study suggested that inhibition of nitric oxide synthase (NOS) by the inhibitor L-NAME increased larval sensitivity to Gram-negative bacterial infection, and reduced expression of a Diptericin-LacZ reporter in the fat body (Foley and O’Farrell, 2003). This study reports that the NOS gene was upregulated following infection in hemocytes, and that feeding with an NO donor induced systemic Diptericin expression, pointing to a role for NOS activity in mediating a robust innate immune response to Gram-negative bacteria. However, microarrays monitoring the immune response of larvae or adult flies to systemic or oral infections show that NOS is not an immune inducible gene (Buchon et al., 2009; De Gregorio et al., 2001; Vodovar et al., 2005). Few follow-up studies have explored roles of NOS in activation of the Imd pathway (but see (Brown et al., 2009; Chakrabarti et al., 2012; Dijkers and O’Farrell, 2007; McGettigan et al., 2005)). Use of a viable null NOS mutant and RT-qPCR did not reveal a striking role for NOS in expression of the Diptericin transcript upon natural infection with the Gram-negative bacterium Ecc15 (Supplementary S24). Loss of NOS may have a subtle impact on reporter expression by affecting fat body maturation, consistent with a role for NOS in regulation of the developmental hormone ecdysone (Jaszczak et al., 2015). The role of NOS in the gut to fat-body signaling, if any, requires further study.
Insights from the reproducibility project on the roles of Eiger and Gr28b in immunity
Eiger has roles in melanization but minor effects on survival and AMP expression
A series of results implicating the TNF-related gene eiger in several aspects of Drosophila immune function (Brandt et al., 2004; Mabery and Schneider, 2010; Schneider et al., 2007) have been called into question due to the recent discovery that widely-used eiger mutants bore a secondary null mutation in the gene encoding the phagocytic receptor NimC1 (Kodra et al., 2020), which has independent roles in immunity (Melcarne et al., 2019). Experiments for ReproSci using clean egr1and egr3 mutants with a wild-type NimC1 locus (Kodra et al., 2020), and NimC11mutants for comparison, suggest that both eiger and NimC1 have independent effects on survival to several pathogens, and that the effect of one or both mutations may explain several survival effects previously attributed solely to Eiger. Contrary to previous results, we find that mutation of eiger does not increase tolerance to Salmonella typhimurium infection, and does not generally increase susceptibility to extracellular pathogens (Supplementary S25, consistent with (Geuking et al., 2009)). However, our results support a role for Eiger in hemolymph melanization in both adult flies and larvae consistent with (Bidla et al., 2007) (Supplementary S26). Experiments done in the framework of ReproSci did not reveal any induction of the eiger gene in the fat body, or a role in regulation of antibacterial peptide gene expression as initially described (Supplementary S27). Immune effects of eiger mutation are largely consistent with the reduced melanization observed in these mutants.
Mutation of Gr28b does not produce anorexia, impair melanization, or affect humoral immunity
(Ayres and Schneider, 2009) found that a mutation in the gene encoding the gustatory receptor Gr28b caused constitutive anorexia and affected fly survival to a number of bacteria. Experiments for ReproSci instead suggest that the observed immune phenotypes are largely attributable to reduced melanization linked to the genetic background of the original Gr28b mutant, as an independent Gr28b mutant does not replicate these phenotypes (Supplementary S28). Furthermore, our results and those of (Sang et al., 2019) indicate that Gr28b mutants are not constitutively anorexic, and this result was likely obtained due to comparison to a wild-type line with unusually high feeding rates.
Discussion
With thousands of papers published in the field of Drosophila immunity, it is difficult to summarize all we know of the fly immune system (Westlake et al., 2024). Today, the Drosophila immune system has been extensively characterized and serves as a model for other insects of medical or agricultural interest. The use of combined genetic approaches makes data obtained in the Drosophila model extremely reliable by current scientific standards. However, it is both impossible and undesirable to entirely avoid publication of controversial or irreproducible results, as science is an incremental and exploratory process where progress is made by constant course correction. That being said, contradictory data or claims that are propagated or not clearly refuted can cause a non-negligeable loss of time to scientists in research development. Taking into consideration the relevance of Drosophila immune research beyond the immediate community and the prestige associated with the use of sophisticated genetic approaches, it is valuable to create easily accessible assessments of established claims in the field, the roots of which may be buried deep in past literature.
In this paper, we have retrospectively analyzed the verifiability of claims from 400 articles accepted for publication before 2011 in the field of Drosophila immunity. We restricted our analysis to Drosophila host defense against bacteria and fungi, which the host laboratory has expertise in. It would be interesting to extend this analysis to Drosophila antiviral and anti-parasitoid immunity with collaboration from relevant laboratories. We expect that application of our approach to viruses and parasitoids would be more challenging, as co-evolved pathogens are expected to display more specific mechanisms that suppress the host immune system (Westlake et al., 2024).
Verifiability of claims was assessed either by analysing subsequent articles or through experimental testing, which we performed for 56 claims. This extensive assessment resource has been centralized on a website that can be accessed (https://ReproSci.epfl.ch/). We hope to continue updating this database as currently unchallenged claims are tested. This article summarizes some of the main findings gained through this extensive reproducibility project. We cannot definitively exclude that some claims highlighted here as challenged could in fact be correct, and that our assessment was erroneous. Reproducibility projects are hampered by the fact that verification must be more stringent and convincing than the initial results to be widely accepted (Errington et al., 2021; Goodman et al., 2016; Shiffrin et al., 2018). However, we encourage the community to continue taking care when considering claims that have been challenged by ReproSci, which may be further clarified with future studies. In our analysis, we were unable to reach definitive conclusions on the immune roles of IRC, Duox and NOS, although we suggest that their roles are not as simple as proposed in the initial articles.
Except for the articles on the role of 18-wheeler in immunity (Ligoxygakis et al., 2002; Williams et al., 1997), PGRP-SD upstream of Toll (Bischoff et al., 2004; Iatsenko et al., 2016) and stimulation of the immune response by LPS (Imler et al., 2000; Kaneko et al., 2004; Leulier et al., 2003), few published results in the field of Drosophila immunity have been formally challenged. While many of the challenged claims discussed in this paper are known to be controversial by experts, or have fallen quietly out of current research interests, some are still a source of confusion. This is particularly true for young scientists new to the field of Drosophila immunity or scientists with primary interests outside of Drosophila immunity that use the field as a reference.
Where possible, we have attempted to clarify the causes that led to irreproducible data or conclusions, as they emphasize challenges inherent to the field of Drosophila immunity and may point to methodologies that require cautious use (Table 1, see Supplementary Table 2 for a list of challenged claims). A first major source of irreproducible results is where the genetic background is responsible for part or all the observed effect rather than the tested mutation. This is often due to use of inappropriate controls, for example where the wild-type comparison has a different genetic background than the mutant of interest. An example of this is the finding that WntD acts in the control of the Toll pathway (Gordon et al., 2005b). Here, we obtained the same results as the authors of the claim when using the same mutant lines, but the result does not stand when using an independent mutant of the same gene, indicating the result was likely due to genetic background. Hemocyte numbers may also vary strongly according to genetic background and rearing conditions (Ramond et al., 2020; Sorrentino et al., 2004). This further complicates in vivo or ex vivo phagocytosis assays, which are often difficult to interpret as the effect of single mutations are often small. In contrast to developmental processes which are more tightly constrained, immune parameters are known to vary strongly among wild-type Drosophila lines (Lazzaro et al., 2004), making analysis of subtle effects difficult. This issue can be addressed in several ways: by using isogenized fly stocks with a controlled genetic background (Ryckebusch et al., 2025), by testing the phenotype of a mutation in an alternate background (e.g., over a deficiency) to see if it holds true, by validating loss of function phenotypes with RNAi, and by performing rescue experiments. None of these various ways offer a full guarantee, and caution in interpretation of the results is always required. A range of wild-type lines (Oregon-R, Canton S, etc.) can be tested to evaluate the natural variability of a phenotype and avoid one-to-one comparisons between a single wild-type and mutant that may be misleading. In some cases, natural polymorphisms affecting immune genes can cause apparent effects on gene expression by affecting primer binding, as suggested here for Defensin. Indeed, during this research we found that a few of published primers (Neyen et al. 2014) were not suited for testing AMP expression across genetic backgrounds.

Statistics on methods associated with challenged claims
Where possible, the reasons behind major claims that were challenged were categorized (see full list in Supplementary Table S2). This table shows the likely reasons for major claims to be challenged, and the percentage of claims that were challenged due to that reason. Only one major claim per article was considered, as the same methodological issue often applied to multiple claims from the same article (Nb of articles=49). 1: The data were interpreted in a certain (erroneous) way due to a lack of available context at the time that the article was published, or alternative explanations were not effectively ruled out. 2: The effects observed were not due to a direct effect on immunity but instead due to effects on development of immune tissues or organs, including hemocytes.
A second source of irreproducible claims is linked to the use of in vitro techniques that do not fully reflect results obtained using in vivo genetics approaches. An example is the initial implication of GNBP1 in the sensing of LPS; although this remains an important discovery, subsequent studies revealed that the role of GNBP1 was indirect, acting as an adapter between PGRP-SA and ModSP . Overexpression of genes can produce ectopic or excessive phenotypes (e.g, indicating a role for PGRP-LE in melanization) that are not supported using loss-of function mutations.
A third source of irreproducible claims is linked to the use of RNAi, which had many off-target effects when the technique was first introduced in the early 2000s (Seinen et al., 2011). The claims related to the serine proteases Spheroid, Sphinx and Spirit (Kambris et al., 2006b) and Undertaker (Cuttell et al., 2008b) appear to be examples of this. Newer generations of RNAi lines have greater specificity towards their target genes. Furthermore, use of several independent RNAi constructs for comparison, and inclusion of driver-only and UAS-only controls, helps to avoid off-target and background effects.
A fourth source of irreproducibility is the use of contaminated or nonspecific reagents. The use of Sigma E. coli LPS solution as an immune elicitor has been a source of confusion in innate immune studies in both vertebrates and Drosophila, as effects attributed to LPS were found to be due to peptidoglycans or lipopeptides also present in the solution (Kaneko et al., 2004). Immune roles may also be attributed to genes that affect the development of immune organs, as exemplified by mutations of 18-wheeler that delay fat body maturation (Ligoxygakis et al., 2002). Many mutations can have similar indirect effects on the immune system, as exemplified by genes involved with ecdysone signaling, which has both indirect and direct effects on immunity that are difficult to disentangle.
Irreproducibility could also be due to differences in rearing conditions, such as flipping regimens that impact the microbiota, dietary composition (Lesperance and Broderick, 2021; Sannino and Dobson, 2023), and use of antifungal agents in food. Many fly stocks harbor cryptic endosymbionts such as Wolbachia or viruses such as Nora virus with subtle phenotypes that can regardless affect immune parameters under certain conditions (Hanson and Lemaitre, 2023; Teixeira et al., 2008). Nora virus, which can be present in up to 50% of laboratory fly lines, can significantly reduce lifespan and skew the outcomes of long-term survival studies (Franchet et al., 2025; Habayeb, 2006; Hanson and Lemaitre, 2023). The very high inducibility of AMP genes used to gauge intensity of the immune response can also be a source of confusion. These genes can be induced by 100-1000-fold upon infection, and their fold change is highly dependent on the basal expression level. If basal AMP expression is elevated in a subset of lines due to cryptic infection or differences in microbiota due to flipping frequency or geographic location, apparent differences in induction may simply result from variations in their baseline expression. The variable and high inducibility of AMP genes explains why they are often identified in microarray studies. This can however create a challenge when analyzing gene expression level, as a 5-10 fold induction is minimal for many AMPs. AMP induction can be contextualized by adding unchallenged animals as a negative control and bacteria-challenged animals as a positive control, which cover the range of induction of AMP genes. Selection of appropriate gene readouts and standards are also important (Neyen et al., 2014; Troha and Buchon, 2019). Finally, we observed that a number of challenged claims involve statements that draw parallels with mammalian immune systems. The tendency for granting agencies and journal editors to value Drosophila research only as a model for mammalian immunity insidiously pressures Drosophila studies to be framed in a human-centric way.
In this article we have not discussed a number of claims, that had no clear follow-up studies and remain unchallenged (see Supplementary Table 3 for a list of unchallenged claims). We encourage members of the Drosophila community to test these, or to contribute unpublished data that addresses these claims. Some examples of these include: the regulation of the Toll pathway by the p62 homolog Drosophila Ref(2)P (Avila et al., 2002), the regulation of AMPs independent of Toll and Imd pathways by the transcription factor FOXO (Becker et al., 2010), the biological significance of Imd pathway activation through cleavage of PGRP-LC by bacterial proteases (Schmidt et al., 2008), or the role of PGRP-LE in antimicrobial autophagy against intracellular bacteria (L. monocytogenes, Mycobacterium marinum) (Yano et al., 2008). In these studies and others, further research should clarify if the observed effect can be reproduced, and if the immune effects are direct or indirect.
In conclusion, our study has assessed the reproducibility of many claims published in the field of Drosophila immunity before 2011. Many articles published in this period were simpler in terms of the number of experiments and techniques used than modern studies, allowing for more efficient assessment. Assessing recent articles might be more useful to the community, to reduce the use of irreproducible data in current areas of research. However, this would likely be challenging as we would lose the benefit of hindsight and progress in the field that comes with time. Here we have favored analysis by claim rather than by article, which allows greater nuance and illustrates that articles may contain both reproducible and irreproducible claims. This highlights the fact that many articles have scientific merit, despite containing challenged claims.
One important goal of this project was to make assessments of claims mainly debated in closed circles more widely accessible, and prevent time loss for scientists who may base research on claims that are widely (but quietly) considered irreproducible. We hope that this effort can assist our community by focusing attention on more promising areas of research, by providing a retrospective on foundational findings in the field, and by identifying common sources of irreproducibility that can be controlled for in the future. In the framework of this project, we opened a web interface that allows the community to comment on these claims and contribute to their assessment. The program used to generate the database is open access, and could eventually be used by other communities to centralize post-publication assessments of articles by claims across research fields.
While we recognize that our approach has subjective elements (identification of claims, choice of claims to test experimentally), we hope that the community-accessible website will allow researchers to voice their opinions and contribute data from many sources to improve objectivity. Our intent is to honor the efforts of the Drosophila community while providing a complement that combats the bias against publishing negative results, which can regardless be essential and informative. A companion article (Lemaitre et al., 2025) discusses the metascience aspect of this project by analyzing patterns of irreproducibility and digging into some sociological reasons behind the publication of erroneous data.
Methods
Generation of the list of annotated articles
The purpose of the ReproSci reproducibility project is to determine whether a wide range of claims published in the field of Drosophila immunity are verifiable using independent sources. A list of publications was generated using a curated search string on the publicly available PubMed database. This search string (Box1) included keywords, MeSH terms and prominent authors in the Drosophila immunity field, and generated an initial list of approximately 2000 publications. This pool was then manually curated to remove 1) reviews and opinion pieces, 2) topics unrelated to Drosophila immunity captured by the search string, and 3) immune topics outside the scope of Lemaitre lab expertise, including development, cell biology, immunity to viruses, immunity to parasitoid wasps, and immunity in other species. Select articles not captured by the search string but known and considered to be key articles in the field were manually added to this selection. The list was then restricted to the date range of interest (1940-2011). This limitation was imposed to allow a time frame in which follow-up studies may have been published that may verify, expand on or contradict the content of the publications selected for the study. These restrictions resulted in a primary list of 400 articles.
Box 1. Search string used on PubMed to generate the initial pool of publications that was subsequently manually curated
The list of articles delivered by the first search string, those that were removed and added, and the final selected list can be found in Table S1. (((“Drosophila proteins”[mesh] and (“antimicrobial cationic peptides”[mesh] or immunity)) or “Drosophila melanogaster/immunology“[MeSH Terms] or “Drosophila/immunology”[MeSH Terms] or (((dimarcq, jean luc[author]) or (meister, marie[author]) or (reichhart, jean marc[author]) or (ligoxigakis, petros[author]) or (lemaitre, bruno[author]) or (leulier, francois[author]) or (kuraishi, takayuki[author]) or (royet, julien[author]) or (carton, yves[author]) or (hoffmann, ja[author]) or (ferrandon, dominique[author]) or (imler, jean luc[author]) or (lagueux, marie[author]) or (ezekowitz, r alan[author]) or (franc, nathalie c[author]) or (maniatis, tom[author]) or (silverman n[author]) or (perrimon, norbert[author]) or (agaisse, herve[author]) or (cherry, sara[author]) or (medzhitov, ruslan[author]) or (engstrom, ylva[author]) or (boman, hans[author]) or (faye, ingrid[author]) or (steiner, hakan[author]) or (hultmark d[author]) or dushay, mitchell[author]) or (theopold, ulrich[author]) or (hedengren m[author]) or (kimbrell, deborah a[author]) or (anderson k[author]) or (schneider, david[author]) or (dionne, marc[author]) or (wu, louisa[author]) or (steward, ruth[author]) or (govind, shubha[author]) or (clark, andrew[author]) or (schlenke, todd[author]) or (lazzaro, brian[author]) or (unckless, robert[author]) or (wasserman s[author]) or (eldon, elizabeth[author]) or (williams mj[author]) or (kurata, shoichiro[author]) or (banerjee u[author]) or (rizki[author]) or (ashburner m[author]) or (kimbrell da[author]) or (teixeira l[author]) or (jiggins, frank[author]) or (Agaisse H[Author]) or (Boutros M[Author]) or (Rahme LG[Author]) or (Steward R[Author]) or (Lee WJ[Author]) or (Nappi A[Author]) or (Carton Y[Author]) or (Steiner H[Author]) or (Jiggins F[Author]) or (Poirié M[Author]) or (Fauvarque MO[Author]) or (Kraaijeveld K[Author]) or (Godfray HC[Author]) or (Andó I[Author]) or (Watnick PI[Author]) or (Söderhäll K[Author]) or (Chung J[Author]) or (Eleftherianos I[Author]) or (Roeder T[Author]) or (Saleh MC[Author]) or (Andino R[Author]) or (Kubli E[Author]) or (van Rij RP[Author]) or (Kawabata SI[Author]) or (Ip YT[Author]) or (Levine M[Author]) or (Ding SW[Author]) or (Brennan CA[Author]) or (Rämet M[Author]) or (Wood W[Author]) or (Foley E[Author]) or (Gateff E[Author]) or (Fauvarque MO[Author]) or (O’Farrell PH[Author]) or (Matova N[Author]) or (Prévost G[Author]) or (Schoofs L[Author]) or (Pastor-Pareja JC[Author]) or (Contamine D[Author]) or (Pan D[Author]) or (Crozatier M[author]) or (Galko MJ[author]) or (Ayres J[Author])) and Drosophila))
Article annotation
Selected primary articles were annotated by a single researcher (HW) and reviewed by a second researcher (BL) to ensure consistency across the project. The ReproSci online database (https://ReproSci.epfl.ch/) was constructed to organize the annotations and provide an interactive and searchable interface. Claims were extracted through careful reading of the selected primary articles and organized according to their importance within the paper: a single Main claim representing the title claim of the paper or overarching finding; 2-4 Major claims emphasized in the abstract or those contributing to the overarching finding; and 0-4 Minor claims representing other findings or note not emphasized in the abstract. Wording of the claims was paraphrased and structured in an attempt to represent claims made directly by the authors, rather than interpretations made by the reader. Other supporting data such as the methods used in the paper and evidence used to support claims was also summarized.
Claims were then cross-checked with evidence from previous, contemporary and subsequent publications and assigned a verification category (see Box 2). A short statement explaining the evidence and citing sources was written to justify the categorization and provide additional context for the claim; this assessment was linked to each claim and can be viewed on the ReproSci website. Supplementary documents summarizing complex or ambiguous topics were also written where necessary and can be viewed on the ReproSci website.
Box 2. Verification categories used to assess claims


Experimental validation
A subset of claims that lacked follow-up in the published literature were selected for direct experimental reproduction by various authors from different laboratory with relevant expertise. This project is concerned with the accuracy of the conclusions themselves rather than the reliability of methods, so we employed indirect or inferential reproducibility, where experimental procedures that may be different from those originally used to reach a conclusion are used to independently verify it. When completed, a document was generated detailing the experiments, including methods and conclusions, conducted to verify an individual claim, which here are summarized as supplementary information. HW and BL take responsibility for the statements made following experimental validation of claims, and acknowledge that our verification experiments could also be erroneous.
These short research projects were also uploaded to the ReproSci website, and the assessment of the claim was adjusted from ‘Unchallenged’ to ‘Verified/Challenged by reproducibility project’ as appropriate. The ReproSci website was made available to the Drosophila scientific community on July 11th 2023, with an email to encourage community members to comment on annotations and verifications and contribute evidence. Experiments performed by various laboratories in the framework of the ReproSci project are summarized in supplemental documents S1 to S28 with their own materials and methods. Additional experimental validation of claims not highlighted in this article can be found on the ReproSci website.
The ReproSci web interface
A versatile web interface was generated to display the 400 articles with annotations and assessments of the reproducibility of the claims made in these articles. This web interface allows i) easy generation of a defined database of articles including titles, authors, date, abstract; ii) an interface for annotation of each article in terms of major, main and minor claims, as well as fields for methods and additional supporting information, iii) a system that allows each claim or individual evidence to be assessed as verified, unchallenged, challenged, with an associated written assessment including references that were used to assign the claims to a chosen category, iv) outputs that tabulate and enumerate articles and claims according to assessment categories in the database, providing data that allow analysis of multiple parameters of reproducibility, and v) an interactive interface allowing community members to comment on and discuss claims and assessments, including uploading of documents to support their discussion. The implementation of the web interface involved the creation of a Ruby-on-Rails web application associated with a PostgreSQL database to store the data in a structured manner. At least 2 main tables store on one hand the annotations and on the other hand the relationships between them (assessments, evidence, comments). Annotations were indexed on-the-fly with the SOLR search engine, allowing subsequent free text search on them. A large part of Javascript code runs on the client side to assist users while typing annotations. Importantly, the application allows registered users to create new workspaces, each workspace focusing on a research subject. In a workspace, the owner of the project is able to moderate and to assign roles to certain users, such as the ability to add articles, enter annotations and assessments, or comment. Through moderation, community comments may be considered, and assessments updated with new information. The code of this web interface can be found at https://github.com/fabdavid/ReproSci/.

The timeline of the Drosophila immunity ReproSci project.
Acknowledgements
We thank the Bloomington Stock Center in the USA and the Vienna Drosophila Resource Center (VDRC) for fly stocks; Paola Bellosta, Oren Schuldiner, Julien Royet, Michèle Crozatier and many members of the Drosophila community for providing fly strains or wasps; the BioImaging & Optics Platform (BIOP) in EPFL for confocal microscopy and the BioInformatics Competence Center of UNIL-EPFL for data analysis. We thank Neal Silverman for sharing information on Dnase II.This work was supported by Swiss National Science Foundation 310030_189085 (BL) and the open-source program by ETH-Domain’s Open Research Data (ORD) Program (2022).
Additional information
Author contributions
Conceptualization: HW, BL
Methodology: HW, BL
Experimental Investigation: HW, YM, TEdB, AC, CM, TS, MP, ZL, YW, LJ, NP, AD, YT, AK, KK, FA, RH, SC, J-PB; FS, MAH, SR, SK, GR, GL, HJ, LX, FD (Francesca Di Cara), EK, BL
Web interface design and coding: FD (Fabrice David)
Funding acquisition: BL, HW
Project administration: BL Supervision: BL
Writing – original draft: BL, HW
Writing – review & editing: BL, HW, FM
Funding
Swiss National Science Foundation (310030_189085)
ETH-Domain’s Open Research Data Program 2022
Additional files
References
- Quantitative Profiling of Drosophila melanogaster Dscam1 Isoforms Reveals No Changes in Splicing after Bacterial ExposurePLOS One 9:e108660https://doi.org/10.1371/journal.pone.0108660Google Scholar
- The Drosophila Atypical Protein Kinase C-Ref(2)P Complex Constitutes a Conserved Module for Signaling in the Toll PathwayMolecular and Cellular Biology 22:8787–8795https://doi.org/10.1128/MCB.22.24.8787-8795.2002Google Scholar
- The role of anorexia in resistance and tolerance to infections in DrosophilaPLoS Biol 7:e1000150https://doi.org/10.1371/journal.pbio.1000150Google Scholar
- Drosophila as a Genetic Model for HematopoiesisGenetics 211:367–417https://doi.org/10.1534/genetics.118.300223Google Scholar
- The Drosophila peptidoglycan-recognition protein LF interacts with peptidoglycan-recognition protein LC to downregulate the Imd pathwayEMBO Rep 12:327–333https://doi.org/10.1038/embor.2011.19Google Scholar
- FOXO-dependent regulation of innate immune homeostasisNature 463:369–373https://doi.org/10.1038/nature08698Google Scholar
- The DH31/CGRP enteroendocrine peptide triggers intestinal contractions favoring the elimination of opportunistic bacteriaPLoS Pathog 14:e1007279https://doi.org/10.1371/journal.ppat.1007279Google Scholar
- Crystal cell rupture after injury in Drosophila requires the JNK pathway, small GTPases and the TNF homolog EigerJournal of Cell Science 120:1209–1215https://doi.org/10.1242/jcs.03420Google Scholar
- Function of the drosophila pattern-recognition receptor PGRP-SD in the detection of Gram-positive bacteriaNat Immunol 5:1175–1180https://doi.org/10.1038/ni1123Google Scholar
- Peptidoglycan Sensing by the Receptor PGRP-LE in the Drosophila Gut Induces Immune Responses to Infectious Bacteria and Tolerance to MicrobiotaCell Host & Microbe 12:153–165https://doi.org/10.1016/j.chom.2012.06.002Google Scholar
- Sequential activation of signaling pathways during innate immune responses in DrosophilaDev Cell 3:711–722https://doi.org/10.1016/s1534-5807(02)00325-8Google Scholar
- Secreted Bacterial Effectors and Host-Produced Eiger/TNF Drive Death in a Salmonella-Infected Fruit FlyPLoS Biology 2:e418https://doi.org/10.1371/journal.pbio.0020418Google Scholar
- Psidin is required in Drosophila blood cells for both phagocytic degradation and immune activation of the fat bodyCurr Biol 17:67–72https://doi.org/10.1016/j.cub.2006.11.026Google Scholar
- The Nimrod transmembrane receptor Eater is required for hemocyte attachment to the sessile compartment in Drosophila melanogasterBiology Open 4:355–363https://doi.org/10.1242/bio.201410595Google Scholar
- Short-Term Starvation of Immune Deficient Drosophila Improves Survival to Gram-Negative Bacterial InfectionsPLoS ONE 4:e4490https://doi.org/10.1371/journal.pone.0004490Google Scholar
- Evolution of innate immunity: clues from invertebrates via fish to mammalsFrontiers in immunology 5:108394Google Scholar
- Invasive and indigenous microbiota impact intestinal stem cell activity through multiple pathways in DrosophilaGenes & Development 23:2333–2344https://doi.org/10.1101/gad.1827009Google Scholar
- Gut homeostasis in a microbial world: insights from Drosophila melanogasterNature Reviews Microbiology 11:615–626https://doi.org/10.1038/nrmicro3074Google Scholar
- A single modular serine protease integrates signals from pattern-recognition receptors upstream of the Drosophila Toll pathwayProc Natl Acad Sci U S A 106:12442–7https://doi.org/10.1073/pnas.0901924106Google Scholar
- Immunity in Drosophila melanogaster — from microbial recognition to whole-organism physiologyNature Reviews Immunology 14:796–810https://doi.org/10.1038/nri3763Google Scholar
- cGAS-like receptor-mediated immunity: the insect perspectiveCurr Opin Immunol 74:183–189https://doi.org/10.1016/j.coi.2022.01.005Google Scholar
- The proPO-system: pros and cons for its role in invertebrate immunityTrends in Immunology 29:263–271https://doi.org/10.1016/j.it.2008.02.009Google Scholar
- Infection-Induced Host Translational Blockage Inhibits Immune Responses and Epithelial Renewal in the Drosophila GutCell Host & Microbe 12:60–70https://doi.org/10.1016/j.chom.2012.06.001Google Scholar
- A Drosophila pattern recognition receptor contains a peptidoglycan docking groove and unusual L,D-carboxypeptidase activityPLoS Biol 2:E277https://doi.org/10.1371/journal.pbio.0020277Google Scholar
- Elimination of plasmatocytes by targeted apoptosis reveals their role in multiple aspects of the Drosophila immune responseProc Natl Acad Sci U S A 106:9797–802https://doi.org/10.1073/pnas.0903971106Google Scholar
- Requirement for a peptidoglycan recognition protein (PGRP) in Relish activation and antibacterial immune responses in DrosophilaScience 296:359–362Google Scholar
- An Effector Peptide Family Required for Drosophila Toll-Mediated ImmunityPLOS Pathogens 11:e1004876https://doi.org/10.1371/journal.ppat.1004876Google Scholar
- Undertaker, a Drosophila Junctophilin, links Draper-mediated phagocytosis and calcium homeostasisCell 135:524–534https://doi.org/10.1016/j.cell.2008.08.033Google Scholar
- Undertaker, a Drosophila Junctophilin, Links Draper-Mediated Phagocytosis and Calcium HomeostasisCell 135:524–534https://doi.org/10.1016/j.cell.2008.08.033Google Scholar
- The evolution of immune mechanismsJ Exp Zool 306B:496–520https://doi.org/10.1002/jez.b.21102Google Scholar
- The Toll and Imd pathways are the major regulators of the immune response in DrosophilaEmbo J 21:2568–79Google Scholar
- The cGAS–STING pathway as a therapeutic target in inflammatory diseasesNat Rev Immunol 21:548–569https://doi.org/10.1038/s41577-021-00524-zGoogle Scholar
- Genetic Ablation of Drosophila Phagocytes Reveals Their Contribution to Both Development and Resistance to Bacterial InfectionJournal of Innate Immunity 1:322–334https://doi.org/10.1159/000210264Google Scholar
- Cooperative control of Drosophila immune responses by the JNK and NF-kappaB signaling pathwaysEMBO J 25:3068–3077https://doi.org/10.1038/sj.emboj.7601182Google Scholar
- Drosophila Calcineurin Promotes Induction of Innate Immune ResponsesCurrent Biology 17:2087–2093https://doi.org/10.1016/j.cub.2007.11.001Google Scholar
- AgDscam, a Hypervariable Immunoglobulin Domain-Containing Receptor of the Anopheles gambiae Innate Immune SystemPLoS Biology 4:e229https://doi.org/10.1371/journal.pbio.0040229Google Scholar
- TrpA1 Regulates Defecation of Food-Borne Pathogens under the Control of the Duox PathwayPLOS Genetics 12:e1005773https://doi.org/10.1371/journal.pgen.1005773Google Scholar
- Drosophila innate immunity: regional and functional specialization of prophenoloxidasesBMC Biology 13https://doi.org/10.1186/s12915-015-0193-6Google Scholar
- Sensing of “danger signals” and pathogen-associated molecular patterns defines binary signaling pathways “upstream” of TollNature Immunology 9:1165–1170https://doi.org/10.1038/ni.1643Google Scholar
- Challenges for assessing replicability in preclinical cancer biologyeLife 10:e67995https://doi.org/10.7554/eLife.67995Google Scholar
- Draper acts through the JNK pathway to control synchronous engulfment of dying germline cells by follicular epithelial cellsDevelopment 139:4029–4039https://doi.org/10.1242/dev.082776Google Scholar
- Nitric oxide contributes to induction of innate immune responses to gram-negative bacteria in DrosophilaGenes & Dev 17:115–25Google Scholar
- Requirement for Croquemort in Phagocytosis of Apoptotic Cells in DrosophilaScience 284:1991–1994https://doi.org/10.1126/science.284.5422.1991Google Scholar
- Croquemort, a novel Drosophila hemocyte/macrophage receptor that recognizes apoptotic cellsImmunity 4:431–443https://doi.org/10.1016/s1074-7613(00)80410-0Google Scholar
- Nora virus proliferates in dividing intestinal stem cells and sensitizes flies to intestinal infection and oxidative stressbioRxiv https://doi.org/10.1101/2025.01.30.635658Google Scholar
- A Non-Redundant Role for Drosophila Mkk4 and Hemipterous/Mkk7 in TAK1-Mediated Activation of JNKPLoS ONE 4:e7709https://doi.org/10.1371/journal.pone.0007709Google Scholar
- Dual activation of the Drosophila toll pathway by two pattern recognition receptorsScience 302:2126–30Google Scholar
- What does research reproducibility mean?Science Translational Medicine 8:341–341https://doi.org/10.1126/scitranslmed.aaf5027Google Scholar
- Pathogenesis of listeria-infected Drosophila wntD mutants is associated with elevated levels of the novel immunity gene edinPLoS Pathog 4:e1000111https://doi.org/10.1371/journal.ppat.1000111Google Scholar
- WntD is a feedback inhibitor of Dorsal/NF-kappaB in Drosophila development and immunityNature 437:746–749https://doi.org/10.1038/nature04073Google Scholar
- WntD is a feedback inhibitor of Dorsal/NF-κB in Drosophila development and immunityNature 437:746–749https://doi.org/10.1038/nature04073Google Scholar
- Cooperative regulation of the induction of the novel antibacterial Listericin by peptidoglycan recognition protein LE and the JAK-STAT pathwayJ Biol Chem 285:15731–15738https://doi.org/10.1074/jbc.M109.082115Google Scholar
- Dual Detection of Fungal Infections in Drosophila via Recognition of Glucans and Sensing of Virulence FactorsCell 127:1425–1437https://doi.org/10.1016/j.cell.2006.10.046Google Scholar
- The Drosophila immune response against Gram-negative bacteria is mediated by a peptidoglycan recognition proteinNature 416:640–644https://doi.org/10.1038/nature734Google Scholar
- An essential role for calcium flux in phagocytes for apoptotic cell engulfment and the anti-inflammatory responseCell Death Differ 16:1323–1331https://doi.org/10.1038/cdd.2009.55Google Scholar
- The Drosophila CD36 Homologue croquemort Is Required to Maintain Immune and Gut Homeostasis during Development and AgingPLoS Pathog 12:e1005961https://doi.org/10.1371/journal.ppat.1005961Google Scholar
- A direct role for dual oxidase in Drosophila gut immunityScience 310:847–850https://doi.org/10.1126/science.1117311Google Scholar
- An antioxidant system required for host protection against gut infection in DrosophilaDev Cell 8:125–132https://doi.org/10.1016/j.devcel.2004.11.007Google Scholar
- Nora virus, a persistent virus in Drosophila, defines a new picorna-like virus familyJournal of General Virology 87:3045–3051https://doi.org/10.1099/vir.0.81997-0Google Scholar
- Epidermal Cells Are the Primary Phagocytes in the Fragmentation and Clearance of Degenerating Dendrites in DrosophilaNeuron 81:544–560https://doi.org/10.1016/j.neuron.2013.11.021Google Scholar
- Antimicrobial peptides do not directly contribute to aging in Drosophila , but improve lifespan by preventing dysbiosisDisease Models & Mechanisms 16:dmm049965https://doi.org/10.1242/dmm.049965Google Scholar
- New insights on Drosophila antimicrobial peptide function in host defense and beyondCurr Opin Immunol 62:22–30https://doi.org/10.1016/j.coi.2019.11.008Google Scholar
- Santa-maria is a glial phagocytic receptor that acts with SIMU to recognize and engulf apoptotic neuronsCell Rep 44:115201https://doi.org/10.1016/j.celrep.2024.115201Google Scholar
- Cutting Edge: Toll-Like Receptor 4 (TLR4)-Deficient Mice Are Hyporesponsive to Lipopolysaccharide: Evidence for TLR4 as the Lps Gene ProductThe Journal of Immunology 162:3749–3752https://doi.org/10.4049/jimmunol.162.7.3749Google Scholar
- Curly Encodes Dual Oxidase, Which Acts with Heme Peroxidase Curly Su to Shape the Adult Drosophila WingPLOS Genetics 11:e1005625https://doi.org/10.1371/journal.pgen.1005625Google Scholar
- Microbiota-Derived Lactate Activates Production of Reactive Oxygen Species by the Intestinal NADPH Oxidase Nox and Shortens Drosophila LifespanImmunity 49:929–942https://doi.org/10.1016/j.immuni.2018.09.017Google Scholar
- PGRP-SD, an Extracellular Pattern-Recognition Receptor, Enhances Peptidoglycan-Mediated Activation of the Drosophila Imd PathwayImmunity 45:1013–1023https://doi.org/10.1016/j.immuni.2016.10.029Google Scholar
- LPS-induced immune response in DrosophilaJ Endotoxin Res 6:459–62Google Scholar
- A Spätzle-Processing Enzyme Required for Toll Signaling Activation in Drosophila Innate ImmunityDevelopmental Cell 10:45–55https://doi.org/10.1016/j.devcel.2005.11.013Google Scholar
- Nitric Oxide Synthase Regulates Growth Coordination During Drosophila melanogaster Imaginal Disc RegenerationGenetics 200:1219–1228https://doi.org/10.1534/genetics.115.178053Google Scholar
- Symbiotic lactobacilli stimulate gut epithelial proliferation via Nox-mediated generation of reactive oxygen speciesThe EMBO Journal 32:3017–3028https://doi.org/10.1038/emboj.2013.224Google Scholar
- Functional analysis of immune response genes in Drosophila identifies JNK pathway as a regulator of antimicrobial peptide gene expression in S2 cellsMicrobes Infect 7:811–819https://doi.org/10.1016/j.micinf.2005.03.014Google Scholar
- Drosophila immunity: a large-scale in vivo RNAi screen identifies five serine proteases required for Toll activationCurr Biol 16:808–813https://doi.org/10.1016/j.cub.2006.03.020Google Scholar
- Drosophila Immunity: A Large-Scale In Vivo RNAi Screen Identifies Five Serine Proteases Required for Toll ActivationCurrent Biology 16:808–813https://doi.org/10.1016/j.cub.2006.03.020Google Scholar
- Monomeric and polymeric gram-negative peptidoglycan but not purified LPS stimulate the Drosophila IMD pathwayImmunity 20:637–49Google Scholar
- A novel mode of induction of the humoral innate immune response in Drosophila larvaeDisease Models & Mechanisms 10:271–281https://doi.org/10.1242/dmm.027102Google Scholar
- A Three-step Proteolytic Cascade Mediates the Activation of the Peptidoglycan-induced Toll Pathway in an InsectJournal of Biological Chemistry 283:7599–7607https://doi.org/10.1074/jbc.M710216200Google Scholar
- Gram-negative bacteria-binding protein, a pattern recognition receptor for lipopolysaccharide and beta-1,3-glucan that mediates the signaling for the induction of innate immune genes in Drosophila melanogaster cellsJ Biol Chem 275:32721–32727https://doi.org/10.1074/jbc.M003934200Google Scholar
- Inhibitor of apoptosis 2 and TAK1-binding protein are components of the Drosophila Imd pathwayEMBO J 24:3423–3434https://doi.org/10.1038/sj.emboj.7600807Google Scholar
- Eater, a transmembrane protein mediating phagocytosis of bacterial pathogens in DrosophilaCell 123:335–346https://doi.org/10.1016/j.cell.2005.08.034Google Scholar
- Nimrod, a putative phagocytosis receptor with EGF repeats in Drosophila plasmatocytesCurr Biol 17:649–654https://doi.org/10.1016/j.cub.2007.02.041Google Scholar
- Hemese, a hemocyte-specific transmembrane protein, affects the cellular immune response in DrosophilaProc Natl Acad Sci USA 100:2622–2627https://doi.org/10.1073/pnas.0436940100Google Scholar
- Post-embryonic hematopoiesis in DrosophilaDev Biol 230:243–57Google Scholar
- Genetic Basis of Natural Variation in D. melanogaster Antibacterial ImmunityScience 303:1873–1876https://doi.org/10.1126/science.1092447Google Scholar
- The dorsoventral regulatory gene cassette spätzle/Toll/cactus controls the potent antifungal response in Drosophila adultsCell 86:973–983Google Scholar
- A retrospective analysis of 400 publications reveals patterns of irreproducibility across an entire life sciences research fieldBioarchive Google Scholar
- Crystal structure of Drosophila PGRP-SD suggests binding to DAP-type but not lysine-type peptidoglycanMol Immunol 45:2521–2530https://doi.org/10.1016/j.molimm.2008.01.015Google Scholar
- Gut Bacteria Mediate Nutrient Availability in Drosophila DietsApplied and Environmental Microbiology 87Google Scholar
- The Drosophila immune system detects bacteria through specific peptidoglycan recognitionNat Immunol 4:478–484https://doi.org/10.1038/ni922Google Scholar
- Sensing microbial infections in the Drosophila melanogaster genetic model organismImmunogenetics 74:35–62https://doi.org/10.1007/s00251-021-01239-0Google Scholar
- Prevalence of Local Immune Response against Oral Infection in a Drosophila/Pseudomonas Infection ModelPLoS Pathogens 2:e56https://doi.org/10.1371/journal.ppat.0020056Google Scholar
- Critical evaluation of the role of the Toll-like receptor 18-Wheeler in the host defense of DrosophilaEMBO Rep 3:666–73Google Scholar
- Structural basis for preferential recognition of diaminopimelic acid-type peptidoglycan by a subset of peptidoglycan recognition proteinsJ Biol Chem 281:8286–8295https://doi.org/10.1074/jbc.M513030200Google Scholar
- Duox activation in Drosophila Malpighian tubules stimulates intestinal epithelial renewal through a countercurrent flowCell Rep 43:114109https://doi.org/10.1016/j.celrep.2024.114109Google Scholar
- The Drosophila TNF Ortholog Eiger Is Required in the Fat Body for a Robust Immune ResponseJournal of Innate Immunity 2:371–378https://doi.org/10.1159/000315050Google Scholar
- Insect renal tubules constitute a cell-autonomous immune system that protects the organism against bacterial infectionInsect Biochemistry and Molecular Biology 35:741–754https://doi.org/10.1016/j.ibmb.2005.02.017Google Scholar
- Exploring new perspectives in immunologyCell 187:2079–2094https://doi.org/10.1016/j.cell.2024.03.038Google Scholar
- Phagocytosis in Drosophila: From molecules and cellular machinery to physiologyInsect Biochemistry and Molecular Biology 109:1–12https://doi.org/10.1016/j.ibmb.2019.04.002Google Scholar
- Two Nimrod receptors, NimC1 and Eater, synergistically contribute to bacterial phagocytosis in Drosophila melanogasterFebs J https://doi.org/10.1111/febs.14857Google Scholar
- Two Nimrod receptors, NimC1 and Eater, synergistically contribute to bacterial phagocytosis in Drosophila melanogasterThe FEBS Journal https://doi.org/10.1111/febs.14857Google Scholar
- Crbn modulates calcium influx by regulating Orai1 during efferocytosisNat Commun 11:5489https://doi.org/10.1038/s41467-020-19272-0Google Scholar
- Parasitoid wasp venom SERCA regulates Drosophila calcium levels and inhibits cellular immunityProc Natl Acad Sci U S A 110:9427–9432https://doi.org/10.1073/pnas.1222351110Google Scholar
- Activation of the innate immunity in Drosophila by endogenous chromosomal DNA that escaped apoptotic degradationGenes Dev 16:2662–2671https://doi.org/10.1101/gad.1022802Google Scholar
- Methods to study Drosophila immunityMethods 68:116–128https://doi.org/10.1016/j.ymeth.2014.02.023Google Scholar
- Tissue- and Ligand-Specific Sensing of Gram-Negative Infection in Drosophila by PGRP-LC Isoforms and PGRP-LEThe Journal of Immunology 189:1886–1897https://doi.org/10.4049/jimmunol.1201022Google Scholar
- Review of Dscam-mediated immunity in shrimp and other arthropodsDevelopmental & Comparative Immunology 46:129–138https://doi.org/10.1016/j.dci.2014.04.002Google Scholar
- Dscam in immunity: A question of diversity in insects and crustaceansDevelopmental & Comparative Immunology 105:103539https://doi.org/10.1016/j.dci.2019.103539Google Scholar
- Dscam1 promotes blood cell survival in Drosophila melanogaster through a dual role in blood cells and neuronsbioRxiv https://doi.org/10.1101/2020.09.26.314997Google Scholar
- Targeting of TAK1 by the NF-kappa B protein Relish regulates the JNK-mediated immune response in DrosophilaGenes Dev 18:584–594https://doi.org/10.1101/gad.1168104Google Scholar
- Clustering of peptidoglycan recognition protein-SA is required for sensing lysine-type peptidoglycan in insectsProceedings of the National Academy of Sciences 104:6602–6607https://doi.org/10.1073/pnas.0610924104Google Scholar
- Damage sensing by a Nox-Ask1-MKK3-p38 signaling pathway mediates regeneration in the adult Drosophila midgutNat Commun 10:4365https://doi.org/10.1038/s41467-019-12336-wGoogle Scholar
- The serine protease homolog spheroide is involved in sensing of pathogenic Gram-positive bacteriaPLOS One 12:e0188339https://doi.org/10.1371/journal.pone.0188339Google Scholar
- Expression cloning of dSR-CI, a class C macrophage-specific scavenger receptor from Drosophila melanogasterProceedings of the National Academy of Sciences 92:4056–4060https://doi.org/10.1073/pnas.92.9.4056Google Scholar
- In vivo RNAi analysis reveals an unexpected role for GNBP1 in the defense against Gram-positive bacterial infection in Drosophila adultsJ Biol Chem Google Scholar
- The conceptual foundations of innate immunity: Taking stock 30 years laterImmunity :613–31Google Scholar
- Functional genomic analysis of phagocytosis and identification of a Drosophila receptor for E. coliNature 416:644–648https://doi.org/10.1038/nature735Google Scholar
- Drosophila scavenger receptor CI is a pattern recognition receptor for bacteriaImmunity 15:1027–1038https://doi.org/10.1016/s1074-7613(01)00249-7Google Scholar
- The adipokine NimrodB5 regulates peripheral hematopoiesis in DrosophilaFebs J 287:3399–3426https://doi.org/10.1111/febs.15237Google Scholar
- An updated proteomic analysis of Drosophila hemolymph after bacterial infectionbioRxiv https://doi.org/10.1101/2024.12.18.629076Google Scholar
- Layers of immunity: Deconstructing the Drosophila effector responseeLife 14:RP107030https://doi.org/10.7554/eLife.107030.1Google Scholar
- An essential complementary role of NF-κB pathway to microbicidal oxidants in Drosophila gut immunityThe EMBO Journal 25:3693–3701https://doi.org/10.1038/sj.emboj.7601233Google Scholar
- Gustatory receptor 28b is necessary for avoiding saponin in Drosophila melanogasterEMBO reports 20:e47328https://doi.org/10.15252/embr.201847328Google Scholar
- Acetobacter pomorum in the Drosophila gut microbiota buffers against host metabolic impacts of dietary preservative formula and batch variation in dietary yeastApplied and Environmental Microbiology 89:e00165–23https://doi.org/10.1128/aem.00165-23Google Scholar
- Infection-induced proteolysis of PGRP-LC controls the IMD activation and melanization cascades in DrosophilaFASEB J 22:918–929https://doi.org/10.1096/fj.06-7907comGoogle Scholar
- Drosophila eiger Mutants Are Sensitive to Extracellular PathogensPLOS Pathogens 3:e41https://doi.org/10.1371/journal.ppat.0030041Google Scholar
- Reproduction–Immunity Trade-Offs in InsectsAnnu Rev Entomol 61:239–256https://doi.org/10.1146/annurev-ento-010715-023924Google Scholar
- RNAi-induced off-target effects in Drosophila melanogaster: frequencies and solutionsBriefings in Functional Genomics 10:206–214https://doi.org/10.1093/bfgp/elr017Google Scholar
- Scientific progress despite irreproducibility: A seeming paradoxProceedings of the National Academy of Sciences 115:2632–2639https://doi.org/10.1073/pnas.1711786114Google Scholar
- Immune activation of NF-kappaB and JNK requires Drosophila TAK1J Biol Chem 278:48928–48934https://doi.org/10.1074/jbc.M304802200Google Scholar
- Reproductive ImmunityIn:
- Rolff J
- Reynolds S
- Genetic analysis of contributions of dorsal group and JAK-Stat92E pathway genes to larval hemocyte concentration and the egg encapsulation response in DrosophilaGenetics 166:1343–56Google Scholar
- Peptidoglycan molecular requirements allowing detection by the Drosophila immune deficiency pathwayJ Immunol 173:7339–48Google Scholar
- Hemocytes are essential for Drosophila melanogaster post-embryonic development, independent of control of the microbiotaDevelopment 149:dev200286https://doi.org/10.1242/dev.200286Google Scholar
- The role of Drosophila microbiota in gut homeostasis and immunityGut Microbes 15:2208503https://doi.org/10.1080/19490976.2023.2208503Google Scholar
- Overexpression of a pattern-recognition receptor, peptidoglycan-recognition protein-LE, activates imd/relish-mediated antibacterial defense and the prophenoloxidase cascade in Drosophila larvaeProc Natl Acad Sci USA 99:13705–13710https://doi.org/10.1073/pnas.212301199Google Scholar
- Peptidoglycan recognition protein (PGRP)-LE and PGRP-LC act synergistically in Drosophila immunityEMBO J 23:4690–4700https://doi.org/10.1038/sj.emboj.7600466Google Scholar
- The bacterial symbiont Wolbachia induces resistance to RNA viral infections in Drosophila melanogasterPLoS Biol 6:e2https://doi.org/10.1371/journal.pbio.1000002Google Scholar
- The Drosophila clotting system and its messages for mammalsDevelopmental & Comparative Immunology 42:42–46https://doi.org/10.1016/j.dci.2013.03.014Google Scholar
- FlyBase 2.0: the next generationNucleic Acids Research 47:D759–D765https://doi.org/10.1093/nar/gky1003Google Scholar
- Methods for the study of innate immunity in Drosophila melanogasterWiley Interdisciplinary Reviews: Developmental Biology e 344https://doi.org/10.1002/wdev.344Google Scholar
- Comparative transcriptomics reveals CrebA as a novel regulator of infection tolerance in D. melanogasterPLoS Pathog 14:e1006847https://doi.org/10.1371/journal.ppat.1006847Google Scholar
- Rnai: a new technology in the post-genomic sequencing eraJ Neurogenet 15:193–204Google Scholar
- Drosophila phagocytosis - still many unknowns under the surfaceAPMIS 119:651–662https://doi.org/10.1111/j.1600-0463.2011.02792.xGoogle Scholar
- Peptidoglycan recognition protein-SD provides versatility of receptor formation in Drosophila immunityProc Natl Acad Sci U S A 105:11881–6https://doi.org/10.1073/pnas.0710092105Google Scholar
- Sensing of Gram-positive bacteria in Drosophila: GNBP1 is needed to process and present peptidoglycan to PGRP-SAEMBO J 25:5005–5014https://doi.org/10.1038/sj.emboj.7601363Google Scholar
- A mechanistic analysis of bacterial recognition and serine protease cascade initiation in larval hemolymph of Manduca sextaInsect Biochemistry and Molecular Biology 148:103818https://doi.org/10.1016/j.ibmb.2022.103818Google Scholar
- Extensive diversity of Ig-superfamily proteins in the immune system of insectsScience 309:1874–1878https://doi.org/10.1126/science.1116887Google Scholar
- Corpse Engulfment Generates a Molecular Memory that Primes the Macrophage Inflammatory ResponseCell 165:1658–1671https://doi.org/10.1016/j.cell.2016.04.049Google Scholar
- Antiviral Immune Responses to Invertebrate Iridescent Virus 6 in DrosophilaUMassChan https://doi.org/10.13028/M25962Google Scholar
- The Drosophila Immunity HandbookEPFL press Google Scholar
- The 18-wheeler mutation reveals complex antibacterial gene regulation in Drosophila host defenseEMBO J 16:6120–6130https://doi.org/10.1093/emboj/16.20.6120Google Scholar
- Lysosomal Degradation Is Required for Sustained Phagocytosis of Bacteria by MacrophagesCell Host Microbe 21:719–730https://doi.org/10.1016/j.chom.2017.05.002Google Scholar
- Macrophage-derived upd3 cytokine causes impaired glucose homeostasis and reduced lifespan in Drosophila fed a lipid-rich dietImmunity 42:133–144https://doi.org/10.1016/j.immuni.2014.12.023Google Scholar
- The Imaginal Disc Growth Factors 2 and 3 participate in the Drosophila response to nematode infectionParasite Immunology e 12581https://doi.org/10.1111/pim.12581Google Scholar
- Autophagic control of listeria through intracellular innate immune recognition in drosophilaNat Immunol 9:908–916https://doi.org/10.1038/ni.1634Google Scholar
- The dual oxidase gene BdDuox regulates the intestinal bacterial community homeostasis of Bactrocera dorsalisISME J 10:1037–1050https://doi.org/10.1038/ismej.2015.202Google Scholar
Article and author information
Author information
Version history
- Preprint posted:
- Sent for peer review:
- Reviewed Preprint version 1:
Cite all versions
You can cite all versions using the DOI https://doi.org/10.7554/eLife.108404. This DOI represents all versions, and will always resolve to the latest one.
Copyright
This is an open-access article, free of all copyright, and may be freely reproduced, distributed, transmitted, modified, built upon, or otherwise used by anyone for any lawful purpose. The work is made available under the Creative Commons CC0 public domain dedication.
Metrics
- views
- 0
- downloads
- 0
- citations
- 0
Views, downloads and citations are aggregated across all versions of this paper published by eLife.