Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 211
Filtrar
1.
PLoS Biol ; 22(5): e3002299, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38713712

RESUMEN

Activation of immune cells requires the remodeling of cell metabolism in order to support immune function. We study these metabolic changes through the infection of Drosophila larvae by parasitoid wasp. The parasitoid egg is neutralized by differentiating lamellocytes, which encapsulate the egg. A melanization cascade is initiated, producing toxic molecules to destroy the egg while the capsule also protects the host from the toxic reaction. We combined transcriptomics and metabolomics, including 13C-labeled glucose and trehalose tracing, as well as genetic manipulation of sugar metabolism to study changes in metabolism, specifically in Drosophila hemocytes. We found that hemocytes increase the expression of several carbohydrate transporters and accordingly uptake more sugar during infection. These carbohydrates are metabolized by increased glycolysis, associated with lactate production, and cyclic pentose phosphate pathway (PPP), in which glucose-6-phosphate is re-oxidized to maximize NADPH yield. Oxidative PPP is required for lamellocyte differentiation and resistance, as is systemic trehalose metabolism. In addition, fully differentiated lamellocytes use a cytoplasmic form of trehalase to cleave trehalose to glucose and fuel cyclic PPP. Intracellular trehalose metabolism is not required for lamellocyte differentiation, but its down-regulation elevates levels of reactive oxygen species, associated with increased resistance and reduced fitness. Our results suggest that sugar metabolism, and specifically cyclic PPP, within immune cells is important not only to fight infection but also to protect the host from its own immune response and for ensuring fitness of the survivor.


Asunto(s)
Glucosa , Hemocitos , Vía de Pentosa Fosfato , Trehalosa , Animales , Trehalosa/metabolismo , Glucosa/metabolismo , Hemocitos/metabolismo , Larva/metabolismo , Larva/parasitología , Drosophila melanogaster/metabolismo , Drosophila melanogaster/parasitología , Resistencia a la Enfermedad , Glucólisis , Interacciones Huésped-Parásitos , Avispas/metabolismo , Avispas/fisiología , Diferenciación Celular , Drosophila/metabolismo , Drosophila/parasitología
2.
BMC Biol ; 22(1): 89, 2024 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-38644510

RESUMEN

BACKGROUND: Innate immune responses can be activated by pathogen-associated molecular patterns (PAMPs), danger signals released by damaged tissues, or the absence of self-molecules that inhibit immunity. As PAMPs are typically conserved across broad groups of pathogens but absent from the host, it is unclear whether they allow hosts to recognize parasites that are phylogenetically similar to themselves, such as parasitoid wasps infecting insects. RESULTS: Parasitoids must penetrate the cuticle of Drosophila larvae to inject their eggs. In line with previous results, we found that the danger signal of wounding triggers the differentiation of specialized immune cells called lamellocytes. However, using oil droplets to mimic infection by a parasitoid wasp egg, we found that this does not activate the melanization response. This aspect of the immune response also requires exposure to parasite molecules. The unidentified factor enhances the transcriptional response in hemocytes and induces a specific response in the fat body. CONCLUSIONS: We conclude that a combination of danger signals and the recognition of nonself molecules is required to activate Drosophila's immune response against parasitic insects.


Asunto(s)
Hemocitos , Interacciones Huésped-Parásitos , Inmunidad Innata , Avispas , Animales , Avispas/fisiología , Interacciones Huésped-Parásitos/inmunología , Hemocitos/inmunología , Drosophila melanogaster/parasitología , Drosophila melanogaster/inmunología , Drosophila melanogaster/fisiología , Larva/inmunología , Larva/parasitología , Drosophila/parasitología , Drosophila/inmunología
3.
PLoS Pathog ; 17(6): e1009656, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34125872

RESUMEN

The exopolysaccharide galactosaminogalactan (GAG) has been well characterized in Aspergilli, especially the human pathogen Aspergillus fumigatus. It has been found that a five-gene cluster is responsible for GAG biosynthesis in Aspergilli to mediate fungal adherence, biofilm formation, immunosuppression or induction of host immune defences. Herein, we report the presence of the conserved GAG biosynthetic gene cluster in the insect pathogenic fungus Metarhizium robertsii to mediate either similar or unique biological functions. Deletion of the gene cluster disabled fungal ability to produce GAG on germ tubes, mycelia and appressoria. Relative to the wild type strain, null mutant was impaired in topical infection but not injection of insect hosts. We found that GAG production by Metarhizium is partially acetylated and could mediate fungal adherence to hydrophobic insect cuticles, biofilm formation, and penetration of insect cuticles. In particular, it was first confirmed that this exopolymer is responsible for the formation of appressorium mucilage, the essential extracellular matrix formed along with the infection structure differentiation to mediate cell attachment and expression of cuticle degrading enzymes. In contrast to its production during A. fumigatus invasive growth, GAG is not produced on the Metarhizium cells harvested from insect hemocoels; however, the polymer can glue germ tubes into aggregates to form mycelium pellets in liquid culture. The results of this study unravel the biosynthesis and unique function of GAG in a fungal system apart from the aspergilli species.


Asunto(s)
Interacciones Huésped-Parásitos/fisiología , Metarhizium/metabolismo , Metarhizium/patogenicidad , Polisacáridos/metabolismo , Virulencia/fisiología , Animales , Drosophila melanogaster/parasitología , Proteínas Fúngicas/metabolismo , Mariposas Nocturnas/parasitología
4.
J Appl Toxicol ; 42(3): 450-474, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34486762

RESUMEN

The use of graphene and multi-walled carbon nanotubes (MWCNTs) has now become rather common in medical applications as well as several other areas thanks to their useful physicochemical properties. While in vitro testing offers some potential, in vivo research into toxic effects of graphene and MWCNTs could yield much more reliable data. Drosophila melanogaster has recently gained significant popularity as a dynamic eukaryotic model in examining toxicity, genotoxicity, and biological effects of exposure to nanomaterials, including oxidative stress, cellular immune response against two strains (NSRef and G486) of parasitoid wasp (Leptopilina boulardi), phenotypic variations, and locomotor behavior risks. D. melanogaster was used as a model organism in our study to identify the potential risks of exposure to graphene (thickness: 2-18 nm) and MWCNTs in different properties (as pure [OD: 10-20 nm short], modified by amide [NH2 ] [OD: 7-13 nm length: 55 µm], and modified by carboxyl [COOH] [OD: 30-50 nm and length: 0.5-2 µm]) at concentrations ranging from 0.1 to 250 µg/ml. Significant effects were observed at two high doses (100 and 250 µg/ml) of graphene or MWCNTs. This is the first study to report findings of cellular immune response against hematopoiesis and parasitoids, nanogenotoxicity, phenotypic variations, and locomotor behavior in D. melanogaster.


Asunto(s)
Daño del ADN , Drosophila melanogaster/efectos de los fármacos , Grafito/toxicidad , Interacciones Huésped-Parásitos/efectos de los fármacos , Nanotubos de Carbono/toxicidad , Estrés Oxidativo/efectos de los fármacos , Animales , Drosophila melanogaster/inmunología , Drosophila melanogaster/parasitología , Drosophila melanogaster/fisiología , Inmunidad Celular/efectos de los fármacos , Locomoción/efectos de los fármacos , Fenotipo
5.
PLoS Genet ; 15(11): e1008452, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31710597

RESUMEN

Trypanosomatid parasites are causative agents of important human and animal diseases such as sleeping sickness and leishmaniasis. Most trypanosomatids are transmitted to their mammalian hosts by insects, often belonging to Diptera (or true flies). These are called dixenous trypanosomatids since they infect two different hosts, in contrast to those that infect just insects (monoxenous). However, it is still unclear whether dixenous and monoxenous trypanosomatids interact similarly with their insect host, as fly-monoxenous trypanosomatid interaction systems are rarely reported and under-studied-despite being common in nature. Here we present the genome of monoxenous trypanosomatid Herpetomonas muscarum and discuss its transcriptome during in vitro culture and during infection of its natural insect host Drosophila melanogaster. The H. muscarum genome is broadly syntenic with that of human parasite Leishmania major. We also found strong similarities between the H. muscarum transcriptome during fruit fly infection, and those of Leishmania during sand fly infections. Overall this suggests Drosophila-Herpetomonas is a suitable model for less accessible insect-trypanosomatid host-parasite systems such as sand fly-Leishmania.


Asunto(s)
Interacciones Huésped-Parásitos/genética , Leishmania/genética , Psychodidae/parasitología , Trypanosomatina/genética , Animales , Drosophila melanogaster/genética , Drosophila melanogaster/parasitología , Infecciones por Euglenozoos/genética , Infecciones por Euglenozoos/parasitología , Infecciones por Euglenozoos/transmisión , Humanos , Insectos Vectores/genética , Leishmania/patogenicidad , Leishmaniasis/genética , Leishmaniasis/parasitología , Leishmaniasis/transmisión , Psychodidae/genética , Trypanosomatina/patogenicidad
6.
PLoS Genet ; 15(3): e1008005, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30875383

RESUMEN

Dipteran or "true" flies occupy nearly every terrestrial habitat, and have evolved to feed upon a wide variety of sources including fruit, pollen, decomposing animal matter, and even vertebrate blood. Here we analyze the molecular, genetic and cellular basis of odor response in the tsetse fly Glossina morsitans, which feeds on the blood of humans and their livestock, and is a vector of deadly trypanosomes. The G. morsitans antenna contains specialized subtypes of sensilla, some of which line a sensory pit not found in the fruit fly Drosophila. We characterize distinct patterns of G. morsitans Odor receptor (GmmOr) gene expression in the antenna. We devise a new version of the "empty neuron" heterologous expression system, and use it to functionally express several GmmOrs in a mutant olfactory receptor neuron (ORN) of Drosophila. GmmOr35 responds to 1-hexen-3-ol, an odorant found in human emanations, and also alpha-pinene, a compound produced by malarial parasites. Another receptor, GmmOr9, which is expressed in the sensory pit, responds to acetone, 2-butanone and 2-propanol. We confirm by electrophysiological recording that neurons of the sensory pit respond to these odorants. Acetone and 2-butanone are strong attractants long used in the field to trap tsetse. We find that 2-propanol is also an attractant for both G. morsitans and the related species G. fuscipes, a major vector of African sleeping sickness. The results identify 2-propanol as a candidate for an environmentally friendly and practical tsetse attractant. Taken together, this work characterizes the olfactory system of a highly distinct kind of fly, and it provides an approach to identifying new agents for controlling the fly and the devastating diseases that it carries.


Asunto(s)
Receptores Odorantes/genética , Atractivos Sexuales/genética , Olfato/genética , Tripanosomiasis Africana/genética , 2-Propanol/química , Animales , Drosophila melanogaster/genética , Drosophila melanogaster/parasitología , Humanos , Aceites/química , Neuronas Receptoras Olfatorias/metabolismo , Neuronas Receptoras Olfatorias/parasitología , Atractivos Sexuales/química , Trypanosoma/genética , Trypanosoma/patogenicidad , Tripanosomiasis Africana/parasitología , Moscas Tse-Tse/genética , Moscas Tse-Tse/patogenicidad
7.
PLoS Genet ; 15(3): e1007931, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30822306

RESUMEN

Dipteran insects transmit serious diseases to humans, often in the form of trypanosomatid parasites. To accelerate research in more difficult contexts of dipteran-parasite relationships, we studied the interaction of the model dipteran Drosophila melanogaster and its natural trypanosomatid Herpetomonas muscarum. Parasite infection reduced fecundity but not lifespan in NF-κB/Relish-deficient flies. Gene expression analysis implicated the two NF-κB pathways Toll and Imd as well as STAT signalling. Tissue specific knock-down of key components of these pathways in enterocytes (ECs) and intestinal stem cells (ISCs) influenced initial numbers, infection dynamics and time of clearance. Herpetomonas triggered STAT activation and proliferation of ISCs. Loss of Relish suppressed ISCs, resulting in increased parasite numbers and delayed clearance. Conversely, overexpression of Relish increased ISCs and reduced uptake. Finally, loss of Toll signalling decreased EC numbers and enabled parasite persistence. This network of signalling may represent a general mechanism with which dipteran respond to trypanosomatids.


Asunto(s)
Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Interacciones Huésped-Parásitos/genética , Factores de Transcripción/genética , Trypanosomatina/genética , Animales , Proliferación Celular/genética , Drosophila melanogaster/parasitología , Enterocitos/metabolismo , Enterocitos/parasitología , Fertilidad/genética , Regulación de la Expresión Génica/genética , Humanos , Intestinos/parasitología , Factores de Transcripción STAT/genética , Transducción de Señal/genética , Células Madre/metabolismo , Receptores Toll-Like/genética , Factor de Transcripción ReIA/genética , Trypanosomatina/patogenicidad
8.
PLoS Pathog ; 15(10): e1008084, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31589659

RESUMEN

It is common to find abundant genetic variation in host resistance and parasite infectivity within populations, with the outcome of infection frequently depending on genotype-specific interactions. Underlying these effects are complex immune defenses that are under the control of both host and parasite genes. We have found extensive variation in Drosophila melanogaster's immune response against the parasitoid wasp Leptopilina boulardi. Some aspects of the immune response, such as phenoloxidase activity, are predominantly affected by the host genotype. Some, such as upregulation of the complement-like protein Tep1, are controlled by the parasite genotype. Others, like the differentiation of immune cells called lamellocytes, depend on the specific combination of host and parasite genotypes. These observations illustrate how the outcome of infection depends on independent genetic effects on different aspects of host immunity. As parasite-killing results from the concerted action of different components of the immune response, these observations provide a physiological mechanism to generate phenomena like epistasis and genotype-interactions that underlie models of coevolution.


Asunto(s)
Drosophila melanogaster/inmunología , Drosophila melanogaster/parasitología , Hemocitos/inmunología , Interacciones Huésped-Parásitos , Inmunidad Humoral/inmunología , Avispas/inmunología , Animales , Drosophila melanogaster/genética , Femenino , Genotipo , Hemocitos/parasitología , Masculino , Monofenol Monooxigenasa/metabolismo , Avispas/genética , Avispas/patogenicidad
9.
PLoS Pathog ; 15(5): e1007626, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31042778

RESUMEN

Parasitic helminths release molecular effectors into their hosts and these effectors can directly damage host tissue and modulate host immunity. Excreted/secreted proteins (ESPs) are one category of parasite molecular effectors that are critical to their success within the host. However, most studies of nematode ESPs rely on in vitro stimulation or culture conditions to collect the ESPs, operating under the assumption that in vitro conditions mimic actual in vivo infection. This assumption is rarely if ever validated. Entomopathogenic nematodes (EPNs) are lethal parasites of insects that produce and release toxins into their insect hosts and are a powerful model parasite system. We compared transcriptional profiles of individual Steinernema feltiae nematodes at different time points of activation under in vitro and in vivo conditions and found that some but not all time points during in vitro parasite activation have similar transcriptional profiles with nematodes from in vivo infections. These findings highlight the importance of experimental validation of ESP collection conditions. Additionally, we found that a suite of genes in the neuropeptide pathway were downregulated as nematodes activated and infection progressed in vivo, suggesting that these genes are involved in host-seeking behavior and are less important during active infection. We then characterized the ESPs of activated S. feltiae infective juveniles (IJs) using mass spectrometry and identified 266 proteins that are released by these nematodes. In comparing these ESPs with those previously identified in activated S. carpocapsae IJs, we identified a core set of 52 proteins that are conserved and present in the ESPs of activated IJs of both species. These core venom proteins include both tissue-damaging and immune-modulating proteins, suggesting that the ESPs of these parasites include both a core set of effectors as well as a specialized set, more adapted to the particular hosts they infect.


Asunto(s)
Drosophila melanogaster/metabolismo , Proteínas del Helminto/metabolismo , Interacciones Huésped-Parásitos , Lepidópteros/metabolismo , Infecciones por Rhabditida/metabolismo , Rabdítidos/patogenicidad , Ponzoñas/metabolismo , Animales , Drosophila melanogaster/parasitología , Perfilación de la Expresión Génica , Proteínas del Helminto/genética , Lepidópteros/parasitología , Infecciones por Rhabditida/parasitología , Simbiosis
10.
PLoS Genet ; 14(7): e1007430, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-30024883

RESUMEN

Many species are able to share information about their environment by communicating through auditory, visual, and olfactory cues. In Drosophila melanogaster, exposure to parasitoid wasps leads to a decline in egg laying, and exposed females communicate this threat to naïve flies, which also depress egg laying. We find that species across the genus Drosophila respond to wasps by egg laying reduction, activate cleaved caspase in oocytes, and communicate the presence of wasps to naïve individuals. Communication within a species and between closely related species is efficient, while more distantly related species exhibit partial communication. Remarkably, partial communication between some species is enhanced after a cohabitation period that requires exchange of visual and olfactory signals. This interspecies "dialect learning" requires neuronal cAMP signaling in the mushroom body, suggesting neuronal plasticity facilitates dialect learning and memory. These observations establish Drosophila as genetic models for interspecies social communication and evolution of dialects.


Asunto(s)
Comunicación Animal , Drosophila melanogaster/fisiología , Interacciones Huésped-Parásitos/fisiología , Cuerpos Pedunculados/metabolismo , Avispas/fisiología , Animales , Caspasas/metabolismo , AMP Cíclico/metabolismo , Drosophila melanogaster/parasitología , Femenino , Masculino , Memoria/fisiología , Cuerpos Pedunculados/fisiología , Plasticidad Neuronal/fisiología , Oocitos/metabolismo , Oviposición/fisiología , Olfato/fisiología
11.
Genomics ; 112(2): 1096-1104, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31247332

RESUMEN

Hymenopteran parasitoid wasps are a diverse collection of species that infect arthropod hosts and use factors found in their venoms to manipulate host immune responses, physiology, and behaviour. Whole parasitoid venoms have been profiled using proteomic approaches, and here we present a bioinformatic characterization of the venom protein content from Ganaspis sp. 1, a parasitoid that infects flies of the genus Drosophila. We find evidence that diverse evolutionary processes including multifunctionalization, co-option, gene duplication, and horizontal gene transfer may be acting in concert to drive venom gene evolution in Ganaspis sp.1. One major role of parasitoid wasp venom is host immune evasion. We previously demonstrated that Ganaspis sp. 1 venom inhibits immune cell activation in infected Drosophila melanogaster hosts, and our current analysis has uncovered additional predicted virulence functions. Overall, this analysis represents an important step towards understanding the composition and activity of parasitoid wasp venoms.


Asunto(s)
Venenos de Artrópodos/genética , Evolución Molecular , Avispas/genética , Animales , Venenos de Artrópodos/metabolismo , Drosophila melanogaster/inmunología , Drosophila melanogaster/parasitología , Duplicación de Gen , Transferencia de Gen Horizontal , Evasión Inmune , Proteoma/genética , Proteoma/metabolismo , Avispas/patogenicidad
12.
J Evol Biol ; 33(11): 1625-1633, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32964555

RESUMEN

When a parasite attacks an insect, the outcome is commonly modulated by the presence of defensive heritable symbionts residing within the insect host. Previous studies noted markedly different strengths of Spiroplasma-mediated fly survival following attack by the same strain of wasp. One difference between the two studies was the strain of Spiroplasma used. We therefore performed a laboratory experiment to assess whether Spiroplasma-mediated protection depends upon the strain of Spiroplasma. We perform this analysis using the two strains of male-killing Spiroplasma used previously, and examined response to challenge by two strains of Leptopilina boulardi and two strains of Leptopilina heterotoma wasp. We found no evidence Spiroplasma strain affected fly survival following wasp attack. In contrast, analysis of the overall level of protection, including the fecundity of survivors of wasp attack, did indicate the two Spiroplasma strains tested varied in protective efficiency against three of the four wasp strains tested. These data highlight the sensitivity of symbiont-mediated protection phenotypes to laboratory conditions, and the importance of common garden comparison. Our results also indicate that Spiroplasma strains can vary in protective capacity in Drosophila, but these differences may exist in the relative performance of survivors of wasp attack, rather than in survival of attack per se.


Asunto(s)
Drosophila melanogaster/microbiología , Drosophila melanogaster/parasitología , Interacciones Huésped-Parásitos , Spiroplasma/genética , Avispas/fisiología , Animales , Femenino , Especificidad de la Especie , Simbiosis
13.
Heredity (Edinb) ; 124(4): 592-602, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31896821

RESUMEN

The ability of an insect to survive attack by natural enemies can be modulated by the presence of defensive symbionts. Study of aphid-symbiont-enemy interactions has indicated that protection may depend on the interplay of symbiont, host and attacking parasite genotypes. However, the importance of these interactions is poorly understood outside of this model system. Here, we study interactions within a Drosophila model system, in which Spiroplasma protect their host against parasitoid wasps and nematodes. We examine whether the strength of protection conferred by Spiroplasma to its host, Drosophila melanogaster varies with strain of attacking Leptopilina heterotoma wasp. We perform this analysis in the presence and absence of ethanol, an environmental factor that also impacts the outcome of parasitism. We observed that Spiroplasma killed all strains of wasp. However, the protection produced by Spiroplasma following wasp attack depended on wasp strain. A composite measure of protection, including both the chance of the fly surviving attack and the relative fecundity/fertility of the survivors, varied from a <4% positive effect of the symbiont following attack of the fly host by the Lh14 strain of wasp to 21% for the Lh-Fr strain in the absence of ethanol. We also observed that environmental ethanol altered the pattern of protection against wasp strains. These data indicate that the dynamics of the Spiroplasma-Drosophila-wasp tripartite interaction depend upon the genetic diversity within the attacking wasp population, and that prediction of symbiont dynamics in natural systems will thus require analysis across natural enemy genotypes and levels of environmental ethanol.


Asunto(s)
Drosophila melanogaster/microbiología , Spiroplasma , Simbiosis , Avispas , Animales , Drosophila melanogaster/parasitología , Etanol , Genotipo , Spiroplasma/fisiología , Avispas/genética , Avispas/patogenicidad
14.
J Eukaryot Microbiol ; 67(5): 583-592, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32498127

RESUMEN

On their spore surfaces, Microsporidia often develop a canopy of filaments with characteristics of intermediate filaments (IF), as we demonstrated in previous studies on Thelohania sp., Ameson michaelis, and Spraguea lophii. Genomic studies indicate that among invertebrates, lamins that may localize in the cytoplasm or nucleus, are the only known IF type. These IFs can bind to the substrate containing cell adhesion molecules (CAMs) cadherins, associated with ß and γ catenins. The objects of this study were to determine whether microsporidia have CAMs with the attached IFs on their envelopes and to find out if these proteins are provided by the host. An examination was made for localization of lamins and CAMs on the spores of the mentioned above species and Anncaliia algerae, plus in the host animals. Then, we determined whether the spores of A. michaelis and A. algerae could bind vertebrate nuclear lamin onto the spore surface. We also tested transgenic Drosophila melanogaster stocks bearing cadherin-GFP to see whether developing A. algerae parasites in these hosts could acquire host CAMs. The tests were positive for all these experiments. We hypothesize that microsporidia are able to acquire host lamin IFs and cell adhesion catenin-cadherin complexes from the host.


Asunto(s)
Adhesión Celular/fisiología , Drosophila melanogaster/parasitología , Interacciones Huésped-Parásitos , Filamentos Intermedios/química , Laminas/química , Microsporidios/fisiología , Animales , Cadherinas/química , Cateninas/química , Microscopía Electrónica de Transmisión , Microsporidios/ultraestructura
15.
Parasitology ; 147(11): 1196-1205, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32498733

RESUMEN

Parasites cause harm to their hosts and represent pervasive causal agents of natural selection. Understanding host proximate responses during interactions with parasites can help predict which genes and molecular pathways are targets of this selection. In the current study, we examined transcriptional changes arising from interactions between Drosophila melanogaster and their naturally occurring ectoparasitic mite, Gamasodes queenslandicus. Shifts in host transcript levels associated with behavioural avoidance revealed the involvement of genes underlying nutrient metabolism. These genetic responses were reflected in altered body lipid and glycogen levels in the flies. Mite infestation triggered a striking immune response, while male accessory gland protein transcript levels were simultaneously reduced, suggesting a trade-off between host immune responses to parasite challenge and reproduction. Comparison of transcriptional analyses during mite infestation to those during nematode and parasitoid attack identified host genes similarly expressed in flies during these interactions. Validation of the involvement of specific genes with RNA interference lines revealed candidates that may directly mediate fly-ectoparasite interactions. Our physiological and molecular characterization of the Drosophila-Gamasodes interface reveals new proximate mechanisms underlying host-parasite interactions, specifically host transcriptional shifts associated with behavioural avoidance and infestation. The results identify potential general mechanisms underlying host resistance and evolutionarily relevant trade-offs.


Asunto(s)
Drosophila melanogaster , Interacciones Huésped-Parásitos , Ácaros , Animales , Evolución Biológica , Drosophila melanogaster/inmunología , Drosophila melanogaster/metabolismo , Drosophila melanogaster/parasitología , Glucógeno/metabolismo , Inmunidad , Metabolismo de los Lípidos , Infestaciones por Ácaros , Parásitos , Reproducción , Transcriptoma
16.
Arch Insect Biochem Physiol ; 105(3): e21736, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32918775

RESUMEN

Parasitic wasps inject various virulence factors into the host insects while laying eggs, among which the venom proteins, one of the key players in host insect/parasitoid relationships, act in host cellular and humoral immune regulation to ensure successful development of wasp progeny. Although the investigations into actions of venom proteins are relatively ample in larval parasitoids, their regulatory mechanisms have not been thoroughly understood in pupal parasitoids. Here, we identified a venom protein, Kazal-type serine protease inhibitor, in the pupal ectoparasitoid Pachycrepoideus vindemiae (PvKazal). Sequence analysis revealed that PvKazal is packed by a signal peptide and a highly conserved "Kazal" domain. Quantitative polymerase chain reaction analysis recorded a higher transcript level of PvKazal in the venom apparatus relative to that in the carcass, and the PvKazal messenger RNA level appeared to reach a peak on day 5 posteclosion. Recombinant PvKazal strongly inhibited the hemolymph melanization of host Drosophila melanogaster. Additionally, the heterologous expression of PvKazal in transgenic Drosophila reduced the crystal cell numbers and blocked the melanization of host pupal hemolymph. Our present work underlying the roles of PvKazal undoubtedly increases the understanding of venom-mediated host-parasitoid crosstalk.


Asunto(s)
Drosophila melanogaster/parasitología , Inhibidores de Serinpeptidasas Tipo Kazal/farmacología , Venenos de Avispas/farmacología , Animales , Drosophila melanogaster/efectos de los fármacos , Hemolinfa/inmunología , Interacciones Huésped-Parásitos , Proteínas de Insectos/química , Proteínas de Insectos/farmacología , Pupa/efectos de los fármacos , Pupa/parasitología , ARN Mensajero , Inhibidores de Serinpeptidasas Tipo Kazal/química , Venenos de Avispas/química , Avispas
17.
Immunogenetics ; 71(7): 501-510, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31147740

RESUMEN

The common fruit fly Drosophila melanogaster is a powerful model for studying signaling pathway regulation. Conserved signaling pathways underlying physiological processes signify evolutionary relationship between organisms and the nature of the mechanisms they control. This study explores the cross-talk between the well-characterized nuclear factor kappa B (NF-κB) innate immune signaling pathways and transforming growth factor beta (TGF-ß) signaling pathway in response to parasitic nematode infection in Drosophila. To understand the link between signaling pathways, we followed on our previous studies by performing a transcript-level analysis of different TGF-ß signaling components following infection of immune-compromised Drosophila adult flies with the nematode parasites Heterorhabditis gerrardi and H. bacteriophora. Our findings demonstrate the requirement of NF-κB transcription factors for activation of TGF-ß signaling pathway in Drosophila in the context of parasitic nematode infection. We observe significant decrease in transcript level of glass bottom boat (gbb) and screw (scw), components of the bone morphogenic protein (BMP) branch, as well as Activinß (actß) which is a component of the Activin branch of the TGF-ß signaling pathway. These results are observed only in H. gerrardi nematode-infected flies compared to uninfected control. Also, this significant decrease in transcript level is found only for extracellular ligands. Future research examining the mechanisms regulating the interaction of these signaling pathways could provide further insight into Drosophila anti-nematode immune function against infection with potent parasitic nematodes.


Asunto(s)
Drosophila melanogaster/parasitología , FN-kappa B/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/inmunología , Perfilación de la Expresión Génica , Interacciones Huésped-Parásitos/genética , Interacciones Huésped-Parásitos/inmunología , FN-kappa B/genética , FN-kappa B/inmunología , Nematodos/microbiología , Nematodos/patogenicidad , Transducción de Señal , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/inmunología
18.
PLoS Pathog ; 13(4): e1006302, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28426766

RESUMEN

Entomopathogenic nematodes (EPNs) are unique parasites due to their symbiosis with entomopathogenic bacteria and their ability to kill insect hosts quickly after infection. It is widely believed that EPNs rely on their bacterial partners for killing hosts. Here we disproved this theory by demonstrating that the in vitro activated infective juveniles (IJs) of Steinernema carpocapsae (a well-studied EPN species) release venom proteins that are lethal to several insects including Drosophila melanogaster. We confirmed that the in vitro activation is a good approximation of the in vivo process by comparing the transcriptomes of individual in vitro and in vivo activated IJs. We further analyzed the transcriptomes of non-activated and activated IJs and revealed a dramatic shift in gene expression during IJ activation. We also analyzed the venom proteome using mass spectrometry. Among the 472 venom proteins, proteases and protease inhibitors are especially abundant, and toxin-related proteins such as Shk domain-containing proteins and fatty acid- and retinol-binding proteins are also detected, which are potential candidates for suppressing the host immune system. Many of the venom proteins have conserved orthologs in vertebrate-parasitic nematodes and are differentially expressed during IJ activation, suggesting conserved functions in nematode parasitism. In summary, our findings strongly support a new model that S. carpocapsae and likely other Steinernema EPNs have a more active role in contributing to the pathogenicity of the nematode-bacterium complex than simply relying on their symbiotic bacteria. Furthermore, we propose that EPNs are a good model system for investigating vertebrate- and human-parasitic nematodes, especially regarding the function of excretory/secretory products.


Asunto(s)
Drosophila melanogaster/parasitología , Interacciones Huésped-Parásitos/fisiología , Infecciones por Nematodos/microbiología , Control Biológico de Vectores , Ponzoñas/metabolismo , Xenorhabdus/aislamiento & purificación , Animales , Insectos/metabolismo , Insectos/microbiología , Simbiosis/fisiología
19.
Biochem Biophys Res Commun ; 502(3): 415-421, 2018 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-29856996

RESUMEN

Acute inflammation can cause serious tissue damage and disease in physiologically-challenged organisms. The precise mechanisms leading to these detrimental effects remain to be determined. In this study, we utilize a reproducible means to induce cellular immune activity in Drosophila larvae in response to mechanical stress. That is, forceps squeeze-administered stress induces lamellocytes, a defensive hemocyte type that normally appears in response to wasp infestation of larvae. The posterior signaling center (PSC) is a cellular microenvironment in the larval hematopoietic lymph gland that is vital for lamellocyte induction upon parasitoid attack. However, we found the PSC was not required for mechanical stress-induced lamellocyte production. In addition, we observed that mechanical injury caused a systemic expression of Unpaired3. This cytokine is both necessary and sufficient to activate the cellular immune response to the imposed stress. These findings provide new insights into the communication between injured tissues and immune system induction, using stress-challenged Drosophila larvae as a tractable model system.


Asunto(s)
Drosophila melanogaster/inmunología , Drosophila melanogaster/metabolismo , Animales , Animales Modificados Genéticamente , Microambiente Celular , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/parasitología , Hemocitos/citología , Hemocitos/inmunología , Inmunidad Celular , Quinasas Janus/metabolismo , Larva/inmunología , Larva/metabolismo , Larva/parasitología , Factores de Transcripción STAT/metabolismo , Transducción de Señal , Estrés Mecánico , Factores de Transcripción/metabolismo , Avispas/inmunología , Avispas/patogenicidad
20.
PLoS Pathog ; 12(7): e1005746, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27414410

RESUMEN

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.


Asunto(s)
Proliferación Celular , Transdiferenciación Celular/fisiología , Drosophila melanogaster/fisiología , Drosophila melanogaster/parasitología , Hematopoyesis/fisiología , Hemocitos/citología , Avispas , Animales , Linaje de la Célula , Citometría de Flujo/métodos , Inmunohistoquímica , Larva , Microscopía Confocal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA