Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
1.
Adv Parasitol ; 70: 99-121, 2009.
Article in English | MEDLINE | ID: mdl-19773068

ABSTRACT

The cellular innate immune response of several species of Drosophila terminates with the encasement of large foreign objects within melanotic capsules comprised of several layers of adhering blood cells or hemocytes. This reaction is manifested by various Drosophila hosts in response to infection by endoparasitic wasps (i.e., parasitoids). Creditable assessments of the factor(s) causing, or contributing to, parasite mortality have long been considered as cytotoxic elements certain molecules associated with enzyme-mediated melanogenesis. However, observations that warrant additional or alternative considerations are those documenting parasitoid survival despite melanotic encapsulation, and those where parasitoids are destroyed with no evidence of this host response. Recent studies of the production of some reactive intermediates of oxygen and nitrogen during infection provide a basis for proposing that these molecules constitute important components of the immune arsenal of Drosophila. Studies of the virulence factors injected by female wasps during oviposition that suppress the host response will likely facilitate identification of the cytotoxic molecules as well as the cell-signaling pathways that regulate their synthesis.


Subject(s)
Drosophila/parasitology , Wasps/physiology , Animals , Drosophila/immunology , Hemocytes/physiology , Host-Parasite Interactions/immunology , Immunity, Cellular/physiology , Melanins/physiology , Monophenol Monooxygenase/antagonists & inhibitors , Virulence Factors/physiology
2.
Adv Parasitol ; 70: 147-88, 2009.
Article in English | MEDLINE | ID: mdl-19773070

ABSTRACT

Interactions between Drosophila hosts and parasitoid wasps are among the few examples in which occurrence of intraspecific variation of parasite success has been studied in natural populations. Such variations can originate from three categories of factors: environmental, host and parasitoid factors. Under controlled laboratory conditions, it is possible to focus on the two last categories, and, using specific reference lines, to analyze their respective importance. Parasitoid and host contributions to variations in parasite success have largely been studied in terms of evolutionary and mechanistic aspects in two Drosophila parasitoids, Asobara tabida and, in more details, in Leptopilina boulardi. This chapter focuses on the physiological and molecular aspects of L. boulardi interactions with two Drosophila host species, while most of the evolutionary hypotheses and models are presented in Chapter 11 of Dupas et al.


Subject(s)
Drosophila/parasitology , Wasps/physiology , Animals , Drosophila/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/parasitology , Genetic Variation , Host-Parasite Interactions/genetics , Host-Parasite Interactions/physiology , Immunity, Innate/genetics , Virulence Factors/physiology , Wasp Venoms/metabolism , Wasps/genetics , Wasps/pathogenicity
3.
Adv Parasitol ; 70: 281-95, 2009.
Article in English | MEDLINE | ID: mdl-19773075

ABSTRACT

In this chapter, we describe the geographically widespread genetic fixation of traits involved in Drosophila-parasitoid immune interactions and the situations where such fixation is not observed. We then discuss how the three classes of coevolutionary dynamics that can occur at the local scale (coevolutionary escalation, coevolutionary alternation and coevolutionary polymorphism), the geographic mosaic of selection, and the phylogenetic constraints may explain such evolutionary patterns and drive diversification in the interactions. Most Drosophila parasitoid traits involved in virulence are host-species specific. Directional selection (coevolutionary escalation) on such traits can lead to their fixation or on the contrary maintain their polymorphism if these traits are associated with fitness costs. When hosts targeted by different host-specific virulence systems coexist, fluctuations in selective pressures on these systems, together with the ability of Drosophila parasitoids to select the most susceptible host for parasitization, can lead to coevolutionary alternation. Finally, we discuss the potential for parasitoid diversification in relation with the fact that most observed geographic situations, for different parasitoid clades, correspond to coevolutionary cold spots, due to fixation of virulence in parasitoid taxa.


Subject(s)
Biological Evolution , Drosophila/parasitology , Host-Parasite Interactions/genetics , Wasps/genetics , Animals , Drosophila/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/parasitology , Genetic Variation , Geography , Phylogeny
4.
J Parasitol ; 95(5): 1134-41, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19388790

ABSTRACT

In larvae of Drosophila paramelanica, eggs and larvae of the endoparasitic wasp Leptopilina heterotoma succumb to an effective host reaction that does not involve blood cell-mediated melanotic encapsulation, a response that characterizes cellular immunity in various species of Drosophila and in many insects and other arthropods. A significant increase occurs, however, in the number of lamellocytes, a type of blood cell that functions in encapsulation reactions. The appearance of activated lamellocytes in D. paramelanica is viewed as an early response to infection, one most likely initiated by non-self-recognition processes that similarly function in other wasp-infected Drosophila. However, ensuing cytotoxic responses, about which little is presently known, are not accompanied by melanotic encapsulation in D. paramelanica. Concurrent analyses of the cell-signaling molecule nitric oxide (*NO) revealed significant alterations in the levels of this free radical during the early stages of infection, most notably a dramatic increase immediately upon infection, and precipitous decreases occurring at times when parasites were killed. Injections of a specific inhibitor of nitric oxide synthase (NOS) into the host's body cavity prior to infection significantly increased parasite survival. These observations suggest some involvement of *NO in the host immune response, either in recruiting hemocytes to sites of infection or as a component of the insect's cytotoxic arsenal, given the capacity of the radical to generate toxic molecules through interactions with various intermediates of oxygen and nitrogen.


Subject(s)
Drosophila/metabolism , Drosophila/parasitology , Nitric Oxide/metabolism , Wasps/physiology , Animals , Drosophila/immunology , Electrochemical Techniques , Enzyme Inhibitors/pharmacology , Female , Hemocytes/cytology , Hemocytes/immunology , Hemolymph/cytology , Hemolymph/immunology , Host-Parasite Interactions , Nitric Oxide Synthase/antagonists & inhibitors , omega-N-Methylarginine/pharmacology
5.
Heredity (Edinb) ; 98(1): 21-7, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17035953

ABSTRACT

Coevolutionary arms races between hosts and parasites would not occur without genetic variation for traits involved in the outcome of parasitism. Genetic variations in resistance and virulence have only rarely been described in pairwise host-parasitoid interactions and have never been analysed in multi-species interactions, in contrast to well-characterized plant-pathogen interactions. This paper reports genetic variation in resistance of Drosophila yakuba to the parasitoid wasp Leptopilina boulardi. The genetic basis and geographic distribution of resistance is analysed. On the basis of these and previous findings, we demonstrate that there are different resistance patterns to the parasitoid species L. boulardi in D. melanogaster and D. yakuba, as well as different specificity levels in the parasitoid species, suggesting complex ecological interactions in the field. This first description of resistance-virulence genetic interactions between a parasitoid and its two host species provides empirical data showing that multi-species interactions may greatly influence coevolutionary processes.


Subject(s)
Biological Evolution , Drosophila/genetics , Drosophila/parasitology , Immunity, Innate/genetics , Wasps/genetics , Animals , Genetic Variation , Host-Parasite Interactions , Wasps/pathogenicity
6.
Genet Res ; 88(2): 81-91, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17125583

ABSTRACT

Drosophila melanogaster resistance against the parasitoid wasp Leptopilina boulardi is under the control of a single gene (Rlb), with two alleles, the resistant one being dominant. Using strains bearing deletions, we previously demonstrated that the 55E2-E6; 55F3 region on chromosome 2R is involved in the resistance phenomenon. In this paper, we first restricted the Rlb containing region by mapping at the molecular level the breakpoints of the Df(2R)Pc66, Df(2R)P34 and Df(2R)Pc4 deficiencies, using both chromosomal in situ hybridization and Southern analyses. The resistance gene was localized in a 100 kb fragment, predicted to contain about 10 different genes. Male recombination genetic experiments were then performed, leading to identification of two possible candidates for the Rlb gene. Potential involvement of one of this genes, edl/mae, is discussed.


Subject(s)
Drosophila melanogaster/genetics , Drosophila melanogaster/parasitology , Genes, Insect , Wasps , Animals , Chromosome Mapping , Cosmids/metabolism , Drosophila Proteins/genetics , Genes, Regulator , In Situ Hybridization , Intracellular Signaling Peptides and Proteins/genetics , Larva/genetics , Larva/metabolism , Male , Membrane Proteins/genetics , Models, Genetic , Recombination, Genetic
7.
J Insect Physiol ; 51(2): 197-205, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15749104

ABSTRACT

Avirulent strains of the endoparasitoid Leptopilina boulardi succumb to a blood cell-mediated melanotic encapsulation response in host larvae of Drosophila melanogaster. Virulent wasp strains effectively abrogate the cellular response with substances introduced into the host that specifically target and effectively suppress one or more immune signaling pathways, including elements that control phenoloxidase-mediated melanotic encapsulation. The present study implicates involvement of the Drosophila Toll pathway in cellular innate immunity by regulating the serine protease inhibitor Serpin 27A (Spn27A), which normally functions as a negative regulator of phenoloxidase. The introduction of Spn27A into normally highly immune competent D. melanogaster larvae significantly reduced their ability to form melanotic capsules around eggs of L. boulardi. This study confirms the role of Spn27A in the melanization cascade and establishes that this pathway and associated blood cell responses can be activated by parasitization. The activation of phenoloxidase and the site-specific localization of the ensuing melanotic response are such critical components of the blood cell response that Spn27A and the signaling elements mediating its activity are likely to represent prime targets for immune suppression by L. boulardi.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/parasitology , Hemocytes/immunology , Immune Tolerance/immunology , Immunity, Innate/drug effects , Serpins/metabolism , Wasps/metabolism , Animals , Blood Cell Count , Drosophila Proteins/pharmacology , Drosophila melanogaster/immunology , Female , Immune Tolerance/drug effects , Immunity, Innate/immunology , Lymphatic System/immunology , Melanins/chemistry , Melanins/metabolism , Monophenol Monooxygenase/metabolism , Ovum/cytology , Ovum/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Serpins/pharmacology , Signal Transduction/immunology
8.
Dev Comp Immunol ; 29(1): 9-32, 2005.
Article in English | MEDLINE | ID: mdl-15325520

ABSTRACT

This review summarizes and compares available data on genetic and molecular aspects of resistance in four well-described invertebrate host-parasite systems: snail-schistosome, mosquito-malaria, mosquito-filarial worm, and Drosophila-wasp associations. It underlies that the major components of the immune reaction, such as hemocyte proliferation and/or activation, and production of cytotoxic radicals are common to invertebrate hosts. Identifying genes responsible for naturally occurring resistance will then be helpful to understand the mechanisms of invertebrate immune defenses and to determine how virulence factors are used by parasites to overcome host resistance. Based on these four well-studied models, invertebrate resistance appears as generally determined by one major locus or a few loci, displaying at least partial dominance. Interestingly, specificity of resistance is highly variable and would involve processes other than simple recognition mechanisms. Finally, resistance was shown to be generally costly but is nevertheless observed at high frequencies in many natural populations, suggesting a high potential for host parasite coevolution.


Subject(s)
Host-Parasite Interactions/genetics , Invertebrates/genetics , Invertebrates/parasitology , Animals , Culicidae/genetics , Culicidae/immunology , Culicidae/parasitology , Drosophila/genetics , Drosophila/immunology , Drosophila/parasitology , Host-Parasite Interactions/immunology , Immunity, Innate , Invertebrates/immunology , Snails/genetics , Snails/immunology , Snails/parasitology , Virulence
9.
J Parasitol ; 90(5): 1139-49, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15562616

ABSTRACT

Immune-suppressive factors (ISFs) introduced into larvae of Drosophila melanogaster during infection by virulent endoparasitic wasps effectively block the innate immune response mediated by blood cells (hemocytes) but have little influence on the autoimmune response made by a tumor strain in which the blood cells manifest a similar response but instead target and destroy endogenous tissues. Quantitative hemocyte analyses indicate that ISFs interfere with the immune effector responses downstream of nonself recognition, hemocyte activation and differentiation, because these responses were manifested by tumor hosts, in which the parasitoids developed. The data suggest that once activated to encapsulate aberrant tissues, the target specificity of the autoimmune-activated hemocytes, and the genetic program underlying tumor formation, cannot be blocked by parasitoid-derived ISFs, which effectively inhibit identical hemocyte-mediated responses during parasitization.


Subject(s)
Drosophila melanogaster/immunology , Wasps/physiology , Animals , Autoimmunity , Drosophila melanogaster/genetics , Drosophila melanogaster/parasitology , Hemocytes/physiology , Host-Parasite Interactions/immunology , Immunity, Cellular , Mutation , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/parasitology , Virulence , Wasps/pathogenicity
10.
Genetica ; 120(1-3): 181-94, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15088657

ABSTRACT

Drosophila species are attacked by a number of parasitoid wasps, which constitute an important factor of population regulation. Since Drosophila melanogaster and Drosophila simulans share common parasitoid species, their ecology and evolution can hardly be understood without considering parasitoids. After a short review of data available on Drosophila-parasitoid interactions involving D. melanogaster and D. simulans as hosts, we report field and laboratory experiments investigating the ecological role of Leptopilina parasitoids in Drosophila communities of southern France. Seasonal survey of species abundance shows that strong interspecific interactions occur at both tropic levels. D. simulans progressively replaces D. melanogaster in southern areas suggesting competitive displacement. Parasitoids are responsible for very high Drosophila mortality (up to 90% in some fruits). Field data emphasize the importance of selective pressure that parasitoids exert on Drosophila communities. The two Leptopilina parasites (L. heterotoma and L boulardi) have different local abundances, which vary in time, and they also compete for hosts. We show that parasitoids can mediate the coexistence of D. melanogaster and D. simulans in the laboratory, and thus may contribute to their puzzling coexistence in the field. Conversely, hosts exert selective pressures on parasitoids, and development on either D. melanogaster or D. simulans strongly affects fitness of adult wasps in a temperature-dependent fashion. Local variation in host species abundance and diversity could thus account for the genetic differentiation we observed in one parasitoid species. Despite laboratory studies cannot fully explain complex field situations, it is clear that the ecology and evolution of Drosophila populations and communities, especially D. melanogaster and D. simulans, are strongly constrained by parasitoids, which should receive more attention.


Subject(s)
Drosophila melanogaster/genetics , Drosophila melanogaster/parasitology , Drosophila/genetics , Drosophila/parasitology , Wasps/genetics , Animals , Ecology , France , Genotype , Host-Parasite Interactions , Pressure , Species Specificity , Temperature
11.
J Insect Physiol ; 49(5): 513-22, 2003 May.
Article in English | MEDLINE | ID: mdl-12770630

ABSTRACT

To develop inside their insect hosts, endoparasitoid wasps must either evade or overcome the host's immune system. Several ichneumonid and braconid wasps inject polydnaviruses that display well-studied immune suppressive effects. However, little is known about the strategies of immunoevasion used by other parasitoid families, such as figitid wasps. The present study provides experimental evidence, based on superparasitism and injection experiments, that the figitid species Leptopilina boulardi uses an active mechanism to suppress the Drosophila melanogaster host immune response, i.e. the encapsulation of the parasitoid eggs. The immune suppressive factors are localised in the long gland and reservoir of the female genital tractus, where virus-like particles (VLPs) have been observed. Parasitism experiments using a host tumorous strain indicate that these factors do not destroy host lamellocytes but that they impair the melanisation pathway. Interestingly, they are not susceptible to heating and are not depleted with prolonged oviposition experience, in contrast to observations reported for L. heterotoma, another figitid species. The mechanisms that prevent encapsulation of eggs from L. boulardi and L. heterotoma differ in several respects, suggesting that different physiological strategies of immunosuppression might be used by specialised and generalist parasitoids.


Subject(s)
Drosophila melanogaster/immunology , Polydnaviridae/physiology , Wasps/physiology , Animals , Female , Microscopy, Electron , Wasps/ultrastructure , Wasps/virology
12.
Heredity (Edinb) ; 90(1): 84-9, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12522430

ABSTRACT

Variations observed in parasite virulence and host resistance may be the outcome of coevolutionary processes. Recent theoretical developments have led to a 'geographic mosaic theory' of coevolution according to which there are some localities where reciprocal selection occurs (hot spots) and others where it is strongly reduced (cold spots). Studies of host-parasitoid systems back this up, revealing a geographical variation of traits subjected to antagonistic selection governed by variations in the strength of the ecological interactions. A more detailed analysis of the genetic basis of these geographic variations in a model system -- the interaction between Drosophila melanogaster and its specific parasitoid Leptopilina boulardi -- suggests that cold spots and hot spots are also driven by the amount of genetic variation available for the trait considered. Our approach, based on isolating reference strains, has been found to predict the result of sympatric interactions and it will be helpful in identifying the selective forces responsible for the coevolution. In this model, host resistance to a standardised reference strain is a weak predictor of the outcome of interactions in the field, and the main parameter accounting for the geographic variations is the number of host species available, with less parasitoid virulence towards D. melanogaster being found in areas displaying a more diversified host community.


Subject(s)
Biological Evolution , Drosophila melanogaster/genetics , Immunity, Innate/genetics , Wasps/genetics , Animals , Drosophila melanogaster/parasitology , Wasps/pathogenicity
13.
Immunogenetics ; 52(3-4): 157-64, 2001.
Article in English | MEDLINE | ID: mdl-11220617

ABSTRACT

Host-parasite relationships represent integrating adaptations of considerable complexity involving the host's immune capacity to both recognize and destroy the parasite, and the latter's ability to successfully invade the host and to circumvent its immune response. Compatibility in Drosophila-parasitic wasp (parasitoid) associations has been shown to have a genetic basis, and to be both species and strain specific. Studies using resistant and susceptible strains of Drosophila melanogaster infected with virulent and avirulent strains of the wasp Leptopilina boulardi demonstrate that the success of the host cellular immune response depends on the genetic status of both host and parasitoid. Immunological, physiological, biochemical, and genetic data form the bases of a two-component model proposed here to account for the observed specificity and complexity of two coevolved adaptations, host nonself recognition and parasitoid virulence.


Subject(s)
Drosophila melanogaster/immunology , Drosophila melanogaster/parasitology , Wasps/immunology , Animals , Drosophila melanogaster/genetics , Hemocytes/immunology , Hemocytes/parasitology , Host-Parasite Interactions/genetics , Host-Parasite Interactions/immunology , Immunogenetics , Models, Biological , Virulence , Wasps/genetics , Wasps/pathogenicity , Wasps/physiology
14.
J Insect Physiol ; 47(2): 167-172, 2001 Feb 01.
Article in English | MEDLINE | ID: mdl-11064023

ABSTRACT

Two strains of Drosophila melanogaster (resistant and susceptible) were parasitized by a virulent or avirulent strain of the parasitoid wasp Leptopilina boulardi. The success of encapsulation depends on both the genetic status of the host strain and the genetic status of the parasitoid strain: the immune cellular reaction (capsule) is observed only with the resistant strain-avirulent strain combination. The total numbers of host haemocytes increased in all 4 combinations, suggesting that an immune reaction was triggered in all hosts. Resistant host larvae infected with the virulent or avirulent strains of parasitoid wasp had slightly more haemocytes per mm(3) than did susceptible host larvae at the beginning of the reaction (less than 15 h post-parasitization). This difference disappeared later. Only the virulent parasitoid strain caused the production of a high percentage of altered lamellocytes (from a discoid shape to a bipolar shape), half the total number of lamellocytes are altered. This suggests that the alteration of lamellocyte shape alone is not sufficient to explain the lack of capsule formation seen in resistant hosts parasitized by the virulent strain. Lastly, there were very few altered lamellocytes in resistant or susceptible hosts parasitized by the avirulent parasitoid strain, two combinations in which no capsule was formed. As is now established for Drosophila-parasitoid interactions, virus-like particles contained in the long gland of the female wasp affect the morphology of the lamellocytes. The results presented here are further proof of the action (direct or indirect) of virus like particles of the virulent strain on lamellocytes.

15.
Proc Biol Sci ; 267(1451): 1417-21, 2000 Jul 22.
Article in English | MEDLINE | ID: mdl-10983825

ABSTRACT

Insect hosts can survive infection by parasitoids using the encapsulation phenomenon. In Drosophila melanogaster the abilities to encapsulate the wasp species Leptopilina boulardi and Asobara tabida each involve one major gene. Both resistance genes have been precisely localized on the second chromosome, 35 centimorgans apart. This result clearly demonstrates the involvement of at least two separate genetic systems in Drosophila resistance to parasitoid wasps. The resistance genes to L. boulardi and A. tabida are not clustered as opposed to many plant resistance genes to pathogens cloned to date.


Subject(s)
Drosophila melanogaster/genetics , Genes, Insect , Wasps/physiology , Animals , Chromosome Mapping , Chromosomes , Drosophila melanogaster/parasitology , Genetic Linkage , Recombination, Genetic
16.
Nitric Oxide ; 4(4): 423-30, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10944427

ABSTRACT

The augmented production of nitric oxide (NO) was observed during the hemocyte-mediated melanotic encapsulation responses of Drosophila melanogaster and D. teissieri. When introduced into the hemocoel of D. melanogaster larvae, NO activated the gene encoding the antimicrobial peptide Diptericin. These observations, together with previous studies documenting the production of superoxide anion (O(*-)(2)) and H(2)O(2) in immune-challenged Drosophila, provide evidence that reactive intermediates of both oxygen (ROI) and nitrogen (RNI) constitute a part of the cytotoxic arsenal employed by Drosophila in defense against both microbial pathogens and eukaryotic parasites. These ROI and RNI appear to represent an evolutionarily conserved innate immune response that is mediated by regulatory proteins that are homologous to those of mammalian species.


Subject(s)
Drosophila/immunology , Drosophila/metabolism , Nitric Oxide/metabolism , Animals , Animals, Genetically Modified , Anti-Infective Agents/metabolism , Drosophila/parasitology , Drosophila Proteins , Gene Expression Regulation/drug effects , Hemocytes/metabolism , Hemolymph/enzymology , Immunohistochemistry , Insect Proteins/genetics , Insect Proteins/metabolism , Larva/drug effects , Larva/metabolism , Larva/parasitology , Models, Biological , Nitric Oxide/immunology , Nitric Oxide/pharmacology , Wasps/physiology , beta-Galactosidase/metabolism
17.
Arch Insect Biochem Physiol ; 43(2): 64-71, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10644970

ABSTRACT

We have used a parasitoid wasp Drosophila melanogaster system to investigate the relationship between the humoral and cellular immune responses in insects. Expression of the gene encoding diptericin, an antibacterial peptide in various D. melanogaster strains parasitized by several species of parasitoid wasps, was studied by Northern blot. These strains have the capacity to encapsulate parasitoid eggs. Two strains appeared to produce diptericin mRNA after parasitoid challenge, regardless of their cellular immune reaction to the wasp species. This suggests that a specific genetic factor, or factors, here designated humoral response to parasitoid (hrtp), is present in these two strains of D. melanogaster and is implicated in the expression of the antibacterial gene after parasite infection. This hrtp genetic factor is recessively expressed and located on the second chromosome, suggesting that it is monofactorial. The transgenic strain Dipt.2.2-lacZ:1, in which the transgene is present on the first chromosome, is normally susceptible to the parasitoid wasp. The chromosome bearing the hrtp factor was transferred to this transgenic strain, which then became reactive when triggered by wasp infection. The hrtp factor appears necessary for the activation of diptericin by the parasitoid wasp. No correlation between the cellular immune capacity and the humoral response was observed, suggesting that the two components of insect immunity are regulated independently. Arch.


Subject(s)
Anti-Bacterial Agents/biosynthesis , Drosophila melanogaster/parasitology , Insect Proteins/physiology , Peptides , Wasps , Animals , Animals, Genetically Modified , Antibody Formation , Drosophila Proteins , Drosophila melanogaster/genetics , Gene Expression Regulation , Immunity, Cellular , Insect Proteins/biosynthesis , Insect Proteins/genetics , Wasps/genetics
18.
Genome Res ; 9(5): 471-81, 1999 May.
Article in English | MEDLINE | ID: mdl-10330127

ABSTRACT

Drosophila melanogaster larvae usually react against eggs of the parasitoid wasp Leptopilina boulardi by surrounding them with a multicellular melanotic capsule. The genetic determinism of this response has been studied previously using susceptible (non-capsule-forming) and resistant (capsule-forming) strains. The results suggest that differences in their encapsulation response involve a single gene, resistance to Leptopilina boulardi (Rlb), with two alleles, the resistant one being dominant. Rlb confers specific protection against Leptopilina boulardi and is thus probably involved in parasitoid recognition. Recent studies have localized this gene on the right arm of the second chromosome and our aim was to precisely determine its genetic and molecular location. Using strains bearing deletions, we demonstrated that resistance to Leptopilina boulardi is conferred by the 55C; 55F3 region and that the 55E2-E6; F3 region is particularly involved. A physical map of the 55C; 56A region was then constructed, based on a set of overlapping cosmid and P1 phage clones. Using single and double digests, cross hybridization of restriction fragments, and location of genetically mapped genes and STSs, a complete, five-enzyme restriction map of this 830-kb region was obtained.


Subject(s)
Drosophila Proteins , Drosophila melanogaster/genetics , Drosophila melanogaster/parasitology , Physical Chromosome Mapping , Wasps/pathogenicity , Animals , Cosmids/genetics , Genes, Dominant , In Situ Hybridization , Larva/genetics , Larva/parasitology , Membrane Proteins/genetics
19.
J Hered ; 89(4): 306-11, 1998.
Article in English | MEDLINE | ID: mdl-9703687

ABSTRACT

Encapsulation has evolved as an efficient mechanism whereby an insect host can survive infection by parasitoids This ability is controlled by a major gene in Drosophila melanogaster hosts. The parasitoid Leptopilina boulardi (Hymenoptera Eucoilidae) can suppress the Drosophila immune reaction by injecting viruslike particles. Analysis of Mendelian crosses between strains of L. boulardi of opposite immune suppressive abilities indicated that the trait is controlled by a single chromosomal factor with semidominant effect. We developed a method to test the monogenic hypothesis. The range of possible genotypic values in back-crosses was studied using various progeny that were genotypically homogenous. These could be obtained because of the arrhenotokous mode of reproduction. The progeny groups were divided into two clusters according to the major gene classification and the hypothesis of another unlinked genetic factor was rejected. Lastly, there was a residual progeny effect within the major groups, indicating that minor genes are also present. This study rules out the polygenic effect for a trait governing the interaction between the insect and parasitoid. It demonstrates that the gene-for-gene model commonly found in plant-parasite interactions may also explain natural variations in insect-parasitoid traits.


Subject(s)
Drosophila melanogaster/genetics , Immune Tolerance , Wasps/genetics , Animals , Crosses, Genetic , Drosophila melanogaster/parasitology , Female , Male , Models, Genetic
20.
Behav Genet ; 28(1): 67-73, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9573648

ABSTRACT

The genetic variability of odor-conditioned probing behavior was investigated in a population of Leptopilina boulardi, a parasitoid of Drosophila larvae. Ovipositor probing is the final step of host location, leading to the discovery of host larvae. It can be triggered by an odor previously experienced during an oviposition as a result of associative learning. This study was based on the analysis of female probing performance over two generations of isofemale lines (using both mother-daughter regressions and one-way analysis of variance). Individual performances of the conditioned response to the odor were characterized by (1) the latency (i.e., the time elapsed between the onset of the odor delivery and the start of the probing response), (2) the duration of the first probing phase, and (3) the total probing duration. Results suggested that the variability of two characters, the latency and the duration of the first probing phase, were under a genetic control in the studied population. This work is the first contribution to quantify the genetic component of this variability.


Subject(s)
Appetitive Behavior/physiology , Conditioning, Psychological/physiology , Host-Parasite Interactions/genetics , Orientation/physiology , Oviposition/genetics , Wasps/genetics , Analysis of Variance , Animals , Chi-Square Distribution , Drosophila/parasitology , Female , Fruit , Linear Models , Odorants , Wasps/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...