Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
Add more filters










Publication year range
1.
Cells ; 13(7)2024 Mar 29.
Article in English | MEDLINE | ID: mdl-38607032

ABSTRACT

Coevolution of hosts and their parasites has shaped heterogeneity of effector hemocyte types, providing immune defense reactions with variable effectiveness. In this work, we characterize hemocytes of Drosophila willistoni, a species that has evolved a cellular immune system with extensive variation and a high degree of plasticity. Monoclonal antibodies were raised and used in indirect immunofluorescence experiments to characterize hemocyte subpopulations, follow their functional features and differentiation. Pagocytosis and parasitization assays were used to determine the functional characteristics of hemocyte types. Samples were visualized using confocal and epifluorescence microscopy. We identified a new multinucleated giant hemocyte (MGH) type, which differentiates in the course of the cellular immune response to parasitoids. These cells differentiate in the circulation through nuclear division and cell fusion, and can also be derived from the central hematopoietic organ, the lymph gland. They have a binary function as they take up bacteria by phagocytosis and are involved in the encapsulation and elimination of the parasitoid. Here, we show that, in response to large foreign particles, such as parasitoids, MGHs differentiate, have a binary function and contribute to a highly effective cellular immune response, similar to the foreign body giant cells of vertebrates.


Subject(s)
Drosophila , Parasites , Animals , Cell Differentiation , Phagocytosis , Immunity, Cellular
2.
Front Immunol ; 14: 1322381, 2023.
Article in English | MEDLINE | ID: mdl-38187383

ABSTRACT

Background: Insects have specialized cell types that participate in the elimination of parasites, for instance, the lamellocytes of the broadly studied species Drosophila melanogaster. Other drosophilids, such as Drosophila ananassae and the invasive Zaprionus indianus, have multinucleated giant hemocytes, a syncytium of blood cells that participate in the encapsulation of the eggs or larvae of parasitoid wasps. These cells can be formed by the fusion of hemocytes in circulation or originate from the lymph gland. Their ultrastructure highly resembles that of the mammalian megakaryocytes. Methods: Morphological, protein expressional, and functional features of blood cells were revealed using epifluorescence and confocal microscopy. The respective hemocyte subpopulations were identified using monoclonal antibodies in indirect immunofluorescence assays. Fluorescein isothiocyanate (FITC)-labeled Escherichia coli bacteria were used in phagocytosis tests. Gene expression analysis was performed following mRNA sequencing of blood cells. Results: D. ananassae and Z. indianus encapsulate foreign particles with the involvement of multinucleated giant hemocytes and mount a highly efficient immune response against parasitoid wasps. Morphological, protein expressional, and functional assays of Z. indianus blood cells suggested that these cells could be derived from large plasmatocytes, a unique cell type developing specifically after parasitoid wasp infection. Transcriptomic analysis of blood cells, isolated from naïve and wasp-infected Z. indianus larvae, revealed several differentially expressed genes involved in signal transduction, cell movements, encapsulation of foreign targets, energy production, and melanization, suggesting their role in the anti-parasitoid response. A large number of genes that encode proteins associated with coagulation and wound healing, such as phenoloxidase activity factor-like proteins, fibrinogen-related proteins, lectins, and proteins involved in the differentiation and function of platelets, were constitutively expressed. The remarkable ultrastructural similarities between giant hemocytes and mammalian megakaryocytes, and presence of platelets, and giant cell-derived anucleated fragments at wound sites hint at the involvement of this cell subpopulation in wound healing processes, in addition to participation in the encapsulation reaction. Conclusion: Our observations provide insights into the broad repertoire of blood cell functions required for efficient defense reactions to maintain the homeostasis of the organism. The analysis of the differentiation and function of multinucleated giant hemocytes gives an insight into the diversification of the immune mechanisms.


Subject(s)
Hemocytes , Wasps , Animals , Drosophila melanogaster , Cell Differentiation , Drosophila , Blood Platelets , Mammals
3.
Elife ; 112022 08 03.
Article in English | MEDLINE | ID: mdl-35920811

ABSTRACT

Hemocytes, similar to vertebrate blood cells, play important roles in insect development and immunity, but it is not well understood how they perform their tasks. New technology, in particular single-cell transcriptomic analysis in combination with Drosophila genetics, may now change this picture. This review aims to make sense of recently published data, focusing on Drosophila melanogaster and comparing to data from other drosophilids, the malaria mosquito, Anopheles gambiae, and the silkworm, Bombyx mori. Basically, the new data support the presence of a few major classes of hemocytes: (1) a highly heterogenous and plastic class of professional phagocytes with many functions, called plasmatocytes in Drosophila and granular cells in other insects. (2) A conserved class of cells that control melanin deposition around parasites and wounds, called crystal cells in D. melanogaster, and oenocytoids in other insects. (3) A new class of cells, the primocytes, so far only identified in D. melanogaster. They are related to cells of the so-called posterior signaling center of the larval hematopoietic organ, which controls the hematopoiesis of other hemocytes. (4) Different kinds of specialized cells, like the lamellocytes in D. melanogaster, for the encapsulation of parasites. These cells undergo rapid evolution, and the homology relationships between such cells in different insects are uncertain. Lists of genes expressed in the different hemocyte classes now provide a solid ground for further investigation of function.


Subject(s)
Bombyx , Drosophila , Animals , Drosophila melanogaster/genetics , Hematopoiesis/genetics , Hemocytes , Insecta
4.
J Innate Immun ; 14(4): 335-354, 2022.
Article in English | MEDLINE | ID: mdl-34864742

ABSTRACT

Multinucleated giant hemocytes (MGHs) represent a novel type of blood cell in insects that participate in a highly efficient immune response against parasitoid wasps involving isolation and killing of the parasite. Previously, we showed that circulating MGHs have high motility and the interaction with the parasitoid rapidly triggers encapsulation. However, structural and molecular mechanisms behind these processes remained elusive. Here, we used detailed ultrastructural analysis and live cell imaging of MGHs to study encapsulation in Drosophila ananassae after parasitoid wasp infection. We found dynamic structural changes, mainly driven by the formation of diverse vesicular systems and newly developed complex intracytoplasmic membrane structures, and abundant generation of giant cell exosomes in MGHs. In addition, we used RNA sequencing to study the transcriptomic profile of MGHs and activated plasmatocytes 72 h after infection, as well as the uninduced blood cells. This revealed that differentiation of MGHs was accompanied by broad changes in gene expression. Consistent with the observed structural changes, transcripts related to vesicular function, cytoskeletal organization, and adhesion were enriched in MGHs. In addition, several orphan genes encoding for hemolysin-like proteins, pore-forming toxins of prokaryotic origin, were expressed at high level, which may be important for parasitoid elimination. Our results reveal coordinated molecular and structural changes in the course of MGH differentiation and parasitoid encapsulation, providing a mechanistic model for a powerful innate immune response.


Subject(s)
Hemocytes , Wasps , Animals , Drosophila , Host-Parasite Interactions , Immunity, Innate , Transcriptome , Wasps/genetics
6.
Sci Rep ; 10(1): 19675, 2020 11 12.
Article in English | MEDLINE | ID: mdl-33184473

ABSTRACT

Nora virus, a virus of Drosophila, encapsidates one of the largest single-stranded RNA virus genomes known. Its taxonomic affinity is uncertain as it has a picornavirus-like cassette of enzymes for virus replication, but the capsid structure was at the time for genome publication unknown. By solving the structure of the virus, and through sequence comparison, we clear up this taxonomic ambiguity in the invertebrate RNA virosphere. Despite the lack of detectable similarity in the amino acid sequences, the 2.7 Å resolution cryoEM map showed Nora virus to have T = 1 symmetry with the characteristic capsid protein ß-barrels found in all the viruses in the Picornavirales order. Strikingly, α-helical bundles formed from the extended C-termini of capsid protein VP4B and VP4C protrude from the capsid surface. They are similar to signalling molecule folds and implicated in virus entry. Unlike other viruses of Picornavirales, no intra-pentamer stabilizing annulus was seen, instead the intra-pentamer stability comes from the interaction of VP4C and VP4B N-termini. Finally, intertwining of the N-termini of two-fold symmetry-related VP4A capsid proteins and RNA, provides inter-pentamer stability. Based on its distinct structural elements and the genetic distance to other picorna-like viruses we propose that Nora virus, and a small group of related viruses, should have its own family within the order Picornavirales.


Subject(s)
Capsid/ultrastructure , Picornaviridae/ultrastructure , Receptors, Virus/metabolism , Binding Sites , Biological Evolution , Capsid/metabolism , Capsid/physiology , Cryoelectron Microscopy , Models, Molecular , Phylogeny , Picornaviridae/classification , Picornaviridae/physiology , Protein Stability , RNA, Viral/metabolism
7.
Sci Rep ; 7(1): 15713, 2017 Nov 16.
Article in English | MEDLINE | ID: mdl-29146985

ABSTRACT

We recently found that JAK/STAT signaling in skeletal muscles is important for the immune response of Drosophila larvae against wasp infection, but it was not clear how muscles could affect the immune response. Here we show that insulin signaling is required in muscles, but not in fat body or hemocytes, during larval development for an efficient encapsulation response and for the formation of lamellocytes. This effect requires TOR signaling. We show that muscle tissue affects the immune response by acting as a master regulator of carbohydrate metabolism in the infected animal, via JAK/STAT and insulin signaling in the muscles, and that there is indirect positive feedback between JAK/STAT and insulin signaling in the muscles. Specifically, stimulation of JAK/STAT signaling in the muscles can rescue the deficient immune response when insulin signaling is suppressed. Our results shed new light on the interaction between metabolism, immunity, and tissue communication.


Subject(s)
Carbohydrate Metabolism , Drosophila melanogaster/immunology , Drosophila melanogaster/parasitology , Muscles/immunology , Muscles/metabolism , Wasps/physiology , Animals , Drosophila melanogaster/metabolism , Glycogen/metabolism , Insulin/metabolism , Janus Kinases/metabolism , Muscles/parasitology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , STAT Transcription Factors/metabolism , Signal Transduction
8.
BMC Genomics ; 18(1): 207, 2017 03 02.
Article in English | MEDLINE | ID: mdl-28249569

ABSTRACT

BACKGROUND: Organisms typically face infection by diverse pathogens, and hosts are thought to have developed specific responses to each type of pathogen they encounter. The advent of transcriptomics now makes it possible to test this hypothesis and compare host gene expression responses to multiple pathogens at a genome-wide scale. Here, we performed a meta-analysis of multiple published and new transcriptomes using a newly developed bioinformatics approach that filters genes based on their expression profile across datasets. Thereby, we identified common and unique molecular responses of a model host species, the honey bee (Apis mellifera), to its major pathogens and parasites: the Microsporidia Nosema apis and Nosema ceranae, RNA viruses, and the ectoparasitic mite Varroa destructor, which transmits viruses. RESULTS: We identified a common suite of genes and conserved molecular pathways that respond to all investigated pathogens, a result that suggests a commonality in response mechanisms to diverse pathogens. We found that genes differentially expressed after infection exhibit a higher evolutionary rate than non-differentially expressed genes. Using our new bioinformatics approach, we unveiled additional pathogen-specific responses of honey bees; we found that apoptosis appeared to be an important response following microsporidian infection, while genes from the immune signalling pathways, Toll and Imd, were differentially expressed after Varroa/virus infection. Finally, we applied our bioinformatics approach and generated a gene co-expression network to identify highly connected (hub) genes that may represent important mediators and regulators of anti-pathogen responses. CONCLUSIONS: Our meta-analysis generated a comprehensive overview of the host metabolic and other biological processes that mediate interactions between insects and their pathogens. We identified key host genes and pathways that respond to phylogenetically diverse pathogens, representing an important source for future functional studies as well as offering new routes to identify or generate pathogen resilient honey bee stocks. The statistical and bioinformatics approaches that were developed for this study are broadly applicable to synthesize information across transcriptomic datasets. These approaches will likely have utility in addressing a variety of biological questions.


Subject(s)
Bees/genetics , Host-Pathogen Interactions/genetics , Animals , Bees/microbiology , Bees/parasitology , Bees/virology , Databases, Genetic , Evolution, Molecular , Gene Expression Regulation , Gene Regulatory Networks , Immunity, Innate/genetics , Molecular Sequence Annotation , Nosema/physiology , RNA Viruses/physiology , Varroidae/physiology
10.
PLoS Pathog ; 12(7): e1005746, 2016 07.
Article in English | MEDLINE | ID: mdl-27414410

ABSTRACT

Cellular immune responses require the generation and recruitment of diverse blood cell types that recognize and kill pathogens. In Drosophila melanogaster larvae, immune-inducible lamellocytes participate in recognizing and killing parasitoid wasp eggs. However, the sequence of events required for lamellocyte generation remains controversial. To study the cellular immune system, we developed a flow cytometry approach using in vivo reporters for lamellocytes as well as for plasmatocytes, the main hemocyte type in healthy larvae. We found that two different blood cell lineages, the plasmatocyte and lamellocyte lineages, contribute to the generation of lamellocytes in a demand-adapted hematopoietic process. Plasmatocytes transdifferentiate into lamellocyte-like cells in situ directly on the wasp egg. In parallel, a novel population of infection-induced cells, which we named lamelloblasts, appears in the circulation. Lamelloblasts proliferate vigorously and develop into the major class of circulating lamellocytes. Our data indicate that lamellocyte differentiation upon wasp parasitism is a plastic and dynamic process. Flow cytometry with in vivo hemocyte reporters can be used to study this phenomenon in detail.


Subject(s)
Cell Proliferation , Cell Transdifferentiation/physiology , Drosophila melanogaster/physiology , Drosophila melanogaster/parasitology , Hematopoiesis/physiology , Hemocytes/cytology , Wasps , Animals , Cell Lineage , Flow Cytometry/methods , Immunohistochemistry , Larva , Microscopy, Confocal
11.
PLoS One ; 11(7): e0159473, 2016.
Article in English | MEDLINE | ID: mdl-27467079

ABSTRACT

To understand how Toll signaling controls the activation of a cellular immune response in Drosophila blood cells (hemocytes), we carried out a genetic modifier screen, looking for deletions that suppress or enhance the mobilization of sessile hemocytes by the gain-of-function mutation Toll10b (Tl10b). Here we describe the results from chromosome arm 3R, where five regions strongly suppressed this phenotype. We identified the specific genes immune response deficient 1 (ird1), headcase (hdc) and possibly Rab23 as suppressors, and we studied the role of ird1 in more detail. An ird1 null mutant and a mutant that truncates the N-terminal kinase domain of the encoded Ird1 protein affected the Tl10b phenotype, unlike mutations that affect the C-terminal part of the protein. The ird1 null mutant suppressed mobilization of sessile hemocytes, but enhanced other Tl10b hemocyte phenotypes, like the formation of melanotic nodules and the increased number of circulating hemocytes. ird1 mutants also had blood cell phenotypes on their own. They lacked crystal cells and showed aberrant formation of lamellocytes. ird1 mutant plasmatocytes had a reduced ability to spread on an artificial substrate by forming protrusions, which may explain why they did not go into circulation in response to Toll signaling. The effect of the ird1 mutation depended mainly on ird1 expression in hemocytes, but ird1-dependent effects in other tissues may contribute. Specifically, the Toll receptor was translocated from the cell membrane to intracellular vesicles in the fat body of the ird1 mutant, and Toll signaling was activated in that tissue, partially explaining the Tl10b-like phenotype. As ird1 is otherwise known to control vesicular transport, we conclude that the vesicular transport system may be of particular importance during an immune response.


Subject(s)
Drosophila/genetics , Fat Body/metabolism , Hemocytes/metabolism , Larva/metabolism , Signal Transduction , Animals , Drosophila/growth & development , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Hemocytes/cytology , Mutation , Phenotype
12.
Virus Res ; 223: 20-7, 2016 09 02.
Article in English | MEDLINE | ID: mdl-27329665

ABSTRACT

Nora virus is an enteric virus that causes persistent, non-pathological infection in Drosophila melanogaster. It replicates in the fly gut and is transmitted via the fecal-oral route. Nora virus has a single-stranded positive-sense RNA genome, which is translated in four open reading frames. Reading frame three encodes the VP3 protein, the structure and function of which we have investigated in this work. We have shown that VP3 is a trimer that has an α-helical secondary structure, with a functionally important coiled-coil domain. In order to identify the role of VP3 in the Nora virus life cycle, we constructed VP3-mutants using the cDNA clone of the virus. Our results show that VP3 does not have a role in the actual assembly of the virus particles, but virions that lack VP3 or harbor VP3 with a disrupted coiled coil domain are incapable of transmission via the fecal-oral route. Removing the region downstream of the putative coiled coil appears to have an effect on the fitness of the virus but does not hamper its replication or transmission. We also found that the VP3 protein and particularly the coiled coil domain are crucial for the stability of Nora virus virions when exposed to heat or proteases. Hence, we propose that VP3 is imperative to Nora virus virions as it confers stability to the viral capsid.


Subject(s)
Capsid Proteins/genetics , Capsid Proteins/metabolism , RNA Viruses/physiology , Virion/physiology , Amino Acid Sequence , Capsid Proteins/chemistry , Gene Expression , Mutation , Protein Conformation, alpha-Helical , Protein Domains , Protein Multimerization , RNA Virus Infections/transmission , RNA Virus Infections/virology , Virion/isolation & purification , Virus Assembly
13.
Sci Rep ; 6: 26250, 2016 05 18.
Article in English | MEDLINE | ID: mdl-27189868

ABSTRACT

We have created a transgenic reporter for virus infection, and used it to study Nora virus infection in Drosophila melanogaster. The transgenic construct, Munin, expresses the yeast transcription factor Gal4, tethered to a transmembrane anchor via a linker that can be cleaved by a viral protease. In infected cells, liberated Gal4 will then transcribe any gene that is linked to a promoter with a UAS motif, the target for Gal4 transcription. For instance, infected cells will glow red in the offspring of a cross between the Munin stock and flies with a UAS-RFP(nls) transgene (expressing a red fluorescent protein). In such flies we show that after natural infection, via the faecal-oral route, 5-15% of the midgut cells are infected, but there is little if any infection elsewhere. By contrast, we can detect infection in many other tissues after injection of virus into the body cavity. The same principle could be applied for other viruses and it could also be used to express or suppress any gene of interest in infected cells.


Subject(s)
Drosophila melanogaster/genetics , Drosophila melanogaster/virology , RNA Virus Infections/genetics , Animals , Animals, Genetically Modified , Drosophila Proteins/genetics , Gastrointestinal Tract/virology , Luminescent Proteins/metabolism , Promoter Regions, Genetic , RNA Viruses/enzymology , Transcription Factors/genetics , Transgenes , Red Fluorescent Protein
14.
Fly (Austin) ; 10(3): 115-22, 2016 07 02.
Article in English | MEDLINE | ID: mdl-27116253

ABSTRACT

Several signaling pathways, including the JAK/STAT and Toll pathways, are known to activate blood cells (hemocytes) in Drosophila melanogaster larvae. They are believed to regulate the immune response against infections by parasitoid wasps, such as Leptopilina boulardi, but how these pathways control the hemocytes is not well understood. Here, we discuss the recent discovery that both muscles and fat body take an active part in this response. Parasitoid wasp infection induces Upd2 and Upd3 secretion from hemocytes, leading to JAK/STAT activation mainly in hemocytes and in skeletal muscles. JAK/STAT activation in muscles, but not in hemocytes, is required for an efficient encapsulation of wasp eggs. This suggests that Upd2 and Upd3 are important cytokines, coordinating different tissues for the cellular immune response in Drosophila. In the fat body, Toll signaling initiates a systemic response in which hemocytes are mobilized and activated hemocytes (lamellocytes) are generated. However, the contribution of Toll signaling to the defense against wasps is limited, probably because the wasps inject inhibitors that prevent the activation of the Toll pathway. In conclusion, parasite infection induces a systemic response in Drosophila larvae involving major organ systems and probably the physiology of the entire organism.


Subject(s)
Drosophila melanogaster/immunology , Drosophila melanogaster/parasitology , Hemocytes/immunology , Host-Parasite Interactions/immunology , Signal Transduction , Wasps/immunology , Animals , Drosophila Proteins/immunology , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Immunity, Cellular/immunology , Janus Kinases/immunology , Janus Kinases/metabolism , Larva/immunology , Larva/parasitology , Muscles/immunology , STAT Transcription Factors/immunology , STAT Transcription Factors/metabolism , Transcription Factors/immunology , Transcription Factors/metabolism , Wasps/pathogenicity
15.
EMBO Rep ; 16(12): 1664-72, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26412855

ABSTRACT

The role of JAK/STAT signaling in the cellular immune response of Drosophila is not well understood. Here, we show that parasitoid wasp infection activates JAK/STAT signaling in somatic muscles of the Drosophila larva, triggered by secretion of the cytokines Upd2 and Upd3 from circulating hemocytes. Deletion of upd2 or upd3, but not the related os (upd1) gene, reduced the cellular immune response, and suppression of the JAK/STAT pathway in muscle cells reduced the encapsulation of wasp eggs and the number of circulating lamellocyte effector cells. These results suggest that JAK/STAT signaling in muscles participates in a systemic immune defense against wasp infection.


Subject(s)
Drosophila Proteins/metabolism , Drosophila/immunology , Drosophila/parasitology , Immunity, Cellular , Janus Kinases/metabolism , STAT Transcription Factors/metabolism , Signal Transduction , Animals , Cytokines/metabolism , Drosophila/genetics , Drosophila Proteins/genetics , Hemocytes/immunology , Host-Parasite Interactions , Immunity, Innate , Janus Kinase 1/metabolism , Janus Kinases/genetics , Larva/genetics , Larva/immunology , Larva/parasitology , Muscles/metabolism , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Signal Transduction/immunology , Transcription Factors/metabolism , Wasps
16.
PLoS Pathog ; 11(5): e1004895, 2015 May.
Article in English | MEDLINE | ID: mdl-25965263

ABSTRACT

The cellular immune response against parasitoid wasps in Drosophila involves the activation, mobilization, proliferation and differentiation of different blood cell types. Here, we have assessed the role of Edin (elevated during infection) in the immune response against the parasitoid wasp Leptopilina boulardi in Drosophila melanogaster larvae. The expression of edin was induced within hours after a wasp infection in larval fat bodies. Using tissue-specific RNAi, we show that Edin is an important determinant of the encapsulation response. Although edin expression in the fat body was required for the larvae to mount a normal encapsulation response, it was dispensable in hemocytes. Edin expression in the fat body was not required for lamellocyte differentiation, but it was needed for the increase in plasmatocyte numbers and for the release of sessile hemocytes into the hemolymph. We conclude that edin expression in the fat body affects the outcome of a wasp infection by regulating the increase of plasmatocyte numbers and the mobilization of sessile hemocytes in Drosophila larvae.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Fat Body/metabolism , Host-Parasite Interactions , Up-Regulation , Wasps/immunology , Animals , Animals, Genetically Modified , Biomarkers/metabolism , Crosses, Genetic , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/genetics , Drosophila melanogaster/cytology , Drosophila melanogaster/immunology , Drosophila melanogaster/parasitology , Fat Body/cytology , Fat Body/immunology , Gene Knockdown Techniques , Genes, Reporter , Hematopoiesis, Extramedullary , Hemocytes/cytology , Hemocytes/immunology , Hemocytes/metabolism , Hemolymph/cytology , Hemolymph/immunology , Hemolymph/metabolism , Immunity, Innate , Kinetics , Larva/cytology , Larva/immunology , Larva/metabolism , Larva/parasitology , Ovum/immunology , Ovum/physiology , Parasite Egg Count , RNA Interference , Recombinant Fusion Proteins/metabolism , Wasps/physiology
17.
Elife ; 42015 Mar 12.
Article in English | MEDLINE | ID: mdl-25764304

ABSTRACT

In a niche under the skin in Drosophila larvae, blood cells called plasmatocytes can transform into other classes of blood cell.


Subject(s)
Hemocytes/cytology , Animals , Drosophila melanogaster
18.
PLoS One ; 9(8): e102568, 2014.
Article in English | MEDLINE | ID: mdl-25102059

ABSTRACT

The Toll signaling pathway, first discovered in Drosophila, has a well-established role in immune responses in insects as well as in mammals. In Drosophila, the Toll-dependent induction of antimicrobial peptide production has been intensely studied as a model for innate immune responses in general. Besides this humoral immune response, Toll signaling is also known to activate blood cells in a reaction that is similar to the cellular immune response to parasite infections, but the mechanisms of this response are poorly understood. Here we have studied this response in detail, and found that Toll signaling in several different tissues can activate a cellular immune defense, and that this response does not require Toll signaling in the blood cells themselves. Like in the humoral immune response, we show that Toll signaling in the fat body (analogous to the liver in vertebrates) is of major importance in the Toll-dependent activation of blood cells. However, this Toll-dependent mechanism of blood cell activation contributes very little to the immune response against the parasitoid wasp, Leptopilina boulardi, probably because the wasp is able to suppress Toll induction. Other redundant pathways may be more important in the defense against this pathogen.


Subject(s)
Drosophila Proteins/physiology , Drosophila/immunology , Fat Body/metabolism , Hemocytes/metabolism , Host-Parasite Interactions/immunology , Immunity, Cellular , Larva/immunology , Toll-Like Receptors/physiology , Animals , Drosophila/metabolism , Drosophila/parasitology , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Larva/metabolism , Larva/parasitology , Signal Transduction , Toll-Like Receptors/genetics , Toll-Like Receptors/metabolism , Wasps/physiology
20.
PLoS Pathog ; 8(8): e1002872, 2012.
Article in English | MEDLINE | ID: mdl-22916019

ABSTRACT

RNA interference (RNAi) is a major antiviral pathway that shapes evolution of RNA viruses. We show here that Nora virus, a natural Drosophila pathogen, is both a target and suppressor of RNAi. We detected viral small RNAs with a signature of Dicer-2 dependent small interfering RNAs in Nora virus infected Drosophila. Furthermore, we demonstrate that the Nora virus VP1 protein contains RNAi suppressive activity in vitro and in vivo that enhances pathogenicity of recombinant Sindbis virus in an RNAi dependent manner. Nora virus VP1 and the viral suppressor of RNAi of Cricket paralysis virus (1A) antagonized Argonaute-2 (AGO2) Slicer activity of RNA induced silencing complexes pre-loaded with a methylated single-stranded guide strand. The convergent evolution of AGO2 suppression in two unrelated insect RNA viruses highlights the importance of AGO2 in antiviral defense.


Subject(s)
Argonaute Proteins/metabolism , Drosophila Proteins/metabolism , Evolution, Molecular , Gene Silencing , Insect Viruses/metabolism , RNA Viruses/metabolism , Animals , Argonaute Proteins/genetics , Drosophila Proteins/genetics , Drosophila melanogaster , Insect Viruses/genetics , RNA Viruses/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...