Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 84
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 51(4): 625-637.e3, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31564469

RESUMEN

Preventing aberrant immune responses against the microbiota is essential for the health of the host. Microbiota-shed pathogen-associated molecular patterns translocate from the gut lumen into systemic circulation. Here, we examined the role of hemolymph (insect blood) filtration in regulating systemic responses to microbiota-derived peptidoglycan. Drosophila deficient for the transcription factor Klf15 (Klf15NN) are viable but lack nephrocytes-cells structurally and functionally homologous to the glomerular podocytes of the kidney. We found that Klf15NN flies were more resistant to infection than wild-type (WT) counterparts but exhibited a shortened lifespan. This was associated with constitutive Toll pathway activation triggered by excess peptidoglycan circulating in Klf15NN flies. In WT flies, peptidoglycan was removed from systemic circulation by nephrocytes through endocytosis and subsequent lysosomal degradation. Thus, renal filtration of microbiota-derived peptidoglycan maintains immune homeostasis in Drosophila, a function likely conserved in mammals and potentially relevant to the chronic immune activation seen in settings of impaired blood filtration.


Asunto(s)
Infecciones Bacterianas/inmunología , Tejido Conectivo/fisiología , Drosophila/fisiología , Glomérulos Renales/fisiología , Factores de Transcripción de Tipo Kruppel/genética , Proteínas Nucleares/genética , Podocitos/fisiología , Animales , Animales Modificados Genéticamente , Secreciones Corporales , Proteínas de Drosophila/metabolismo , Endocitosis , Homeostasis , Inmunidad Innata , Mamíferos , Microbiota , Receptores Toll-Like/metabolismo
2.
PLoS Pathog ; 20(8): e1012447, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39133760

RESUMEN

Overnutrition with dietary sugar can worsen infection outcomes in diverse organisms including insects and humans, through generally unknown mechanisms. In the present study, we show that adult Drosophila melanogaster fed high-sugar diets became more susceptible to infection by the Gram-negative bacteria Providencia rettgeri and Serratia marcescens. We found that P. rettgeri and S. marcescens proliferate more rapidly in D. melanogaster fed a high-sugar diet, resulting in increased probability of host death. D. melanogaster become hyperglycemic on the high-sugar diet, and we find evidence that the extra carbon availability may promote S. marcescens growth within the host. However, we found no evidence that increased carbon availability directly supports greater P. rettgeri growth. D. melanogaster on both diets fully induce transcription of antimicrobial peptide (AMP) genes in response to infection, but D. melanogaster provided with high-sugar diets show reduced production of AMP protein. Thus, overnutrition with dietary sugar may impair host immunity at the level of AMP translation. Our results demonstrate that dietary sugar can shape infection dynamics by impacting both host and pathogen, depending on the nutritional requirements of the pathogen and by altering the physiological capacity of the host to sustain an immune response.


Asunto(s)
Drosophila melanogaster , Animales , Drosophila melanogaster/microbiología , Providencia , Serratia marcescens/patogenicidad , Azúcares de la Dieta/efectos adversos , Susceptibilidad a Enfermedades , Infecciones por Serratia/microbiología , Infecciones por Enterobacteriaceae/microbiología , Péptidos Antimicrobianos/metabolismo
3.
PLoS Pathog ; 19(8): e1011588, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37651317

RESUMEN

Several aspects of mosquito ecology that are important for vectored disease transmission and control have been difficult to measure at epidemiologically important scales in the field. In particular, the ability to describe mosquito population structure and movement rates has been hindered by difficulty in quantifying fine-scale genetic variation among populations. The mosquito virome represents a possible avenue for quantifying population structure and movement rates across multiple spatial scales. Mosquito viromes contain a diversity of viruses, including several insect-specific viruses (ISVs) and "core" viruses that have high prevalence across populations. To date, virome studies have focused on viral discovery and have only recently begun examining viral ecology. While nonpathogenic ISVs may be of little public health relevance themselves, they provide a possible route for quantifying mosquito population structure and dynamics. For example, vertically transmitted viruses could behave as a rapidly evolving extension of the host's genome. It should be possible to apply established analytical methods to appropriate viral phylogenies and incidence data to generate novel approaches for estimating mosquito population structure and dispersal over epidemiologically relevant timescales. By studying the virome through the lens of spatial and genomic epidemiology, it may be possible to investigate otherwise cryptic aspects of mosquito ecology. A better understanding of mosquito population structure and dynamics are key for understanding mosquito-borne disease ecology and methods based on ISVs could provide a powerful tool for informing mosquito control programs.


Asunto(s)
Virus de Insectos , Animales , Ecología , Vectores Genéticos , Genómica , Insectos
4.
J Invertebr Pathol ; 204: 108084, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38452853

RESUMEN

Opportunistic bacterial infections are common in insect populations but there is little information on how they are acquired or transmitted. We tested the hypothesis that Macrocheles mites can transmit systemic bacterial infections between Drosophila hosts. We found that 24% of mites acquired detectable levels of bacteria after feeding on infected flies and 87% of infected mites passed bacteria to naïve recipient flies. The probability that a mite could pass Serratia from an infected donor fly to a naïve recipient fly was 27.1%. These data demonstrate that Macrocheles mites are capable of serving as vectors of bacterial infection between insects.


Asunto(s)
Ácaros , Animales , Ácaros/microbiología , Ácaros/fisiología , Drosophila/microbiología , Drosophila/parasitología , Serratia/fisiología , Drosophila melanogaster/microbiología
5.
BMC Biol ; 20(1): 127, 2022 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-35655304

RESUMEN

BACKGROUND: Single tissues can have multiple functions, which can result in constraints, impaired function, and tradeoffs. The insect fat body performs remarkably diverse functions including metabolic control, reproductive provisioning, and systemic immune responses. How polyfunctional tissues simultaneously execute multiple distinct physiological functions is generally unknown. Immunity and reproduction are observed to trade off in many organisms but the mechanistic basis for this tradeoff is also typically not known. Here we investigate constraints and trade-offs in the polyfunctional insect fat body. RESULTS: Using single-nucleus sequencing, we determined that the Drosophila melanogaster fat body executes diverse basal functions with heterogenous cellular subpopulations. The size and identity of these subpopulations are remarkably stable between virgin and mated flies, as well as before and after infection. However, as an emergency function, the immune response engages the entire tissue and all cellular subpopulations produce induce expression of defense genes. We found that reproductively active females who were given bacterial infection exhibited signatures of ER stress and impaired capacity to synthesize new protein in response to infection, including decreased capacity to produce antimicrobial peptides. Transient provision of a reversible translation inhibitor to mated females prior to infection rescued general protein synthesis, specific production of antimicrobial peptides, and survival of infection. CONCLUSIONS: The commonly observed tradeoff between reproduction and immunity appears to be driven, in D. melanogaster, by a failure of the fat body to be able to handle simultaneous protein translation demands of reproductive provisioning and immune defense. We suggest that inherent cellular limitations in tissues that perform multiple functions may provide a general explanation for the wide prevalence of physiological and evolutionary tradeoffs.


Asunto(s)
Drosophila melanogaster , Reproducción , Animales , Drosophila melanogaster/genética , Femenino , Reproducción/genética
6.
Mol Biol Evol ; 38(12): 5782-5805, 2021 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-34469576

RESUMEN

Drosophila melanogaster is a leading model in population genetics and genomics, and a growing number of whole-genome data sets from natural populations of this species have been published over the last years. A major challenge is the integration of disparate data sets, often generated using different sequencing technologies and bioinformatic pipelines, which hampers our ability to address questions about the evolution of this species. Here we address these issues by developing a bioinformatics pipeline that maps pooled sequencing (Pool-Seq) reads from D. melanogaster to a hologenome consisting of fly and symbiont genomes and estimates allele frequencies using either a heuristic (PoolSNP) or a probabilistic variant caller (SNAPE-pooled). We use this pipeline to generate the largest data repository of genomic data available for D. melanogaster to date, encompassing 271 previously published and unpublished population samples from over 100 locations in >20 countries on four continents. Several of these locations have been sampled at different seasons across multiple years. This data set, which we call Drosophila Evolution over Space and Time (DEST), is coupled with sampling and environmental metadata. A web-based genome browser and web portal provide easy access to the SNP data set. We further provide guidelines on how to use Pool-Seq data for model-based demographic inference. Our aim is to provide this scalable platform as a community resource which can be easily extended via future efforts for an even more extensive cosmopolitan data set. Our resource will enable population geneticists to analyze spatiotemporal genetic patterns and evolutionary dynamics of D. melanogaster populations in unprecedented detail.


Asunto(s)
Drosophila melanogaster , Metagenómica , Animales , Drosophila melanogaster/genética , Frecuencia de los Genes , Genética de Población , Genómica
7.
Proc Biol Sci ; 288(1951): 20210786, 2021 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-34034518

RESUMEN

A long-standing question in infection biology is why two very similar individuals, with very similar pathogen exposures, may have very different outcomes. Recent experiments have found that even isogenic Drosophila melanogaster hosts, given identical inoculations of some bacterial pathogens at suitable doses, can experience very similar initial bacteria proliferation but then diverge to either a lethal infection or a sustained chronic infection with much lower pathogen load. We hypothesized that divergent infection outcomes are a natural result of mutual negative feedbacks between pathogens and the host immune response. Here, we test this hypothesis in silico by constructing process-based dynamic models for bacterial population growth, host immune induction and the feedbacks between them, based on common mechanisms of immune system response. Mathematical analysis of a minimal conceptual model confirms our qualitative hypothesis that mutual negative feedbacks can magnify small differences among hosts into life-or-death differences in outcome. However, explaining observed features of chronic infections requires an extension of the model to include induced pathogen modifications that shield themselves from host immune responses at the cost of reduced proliferation rate. Our analysis thus generates new, testable predictions about the mechanisms underlying bimodal infection outcomes.


Asunto(s)
Drosophila melanogaster , Interacciones Huésped-Patógeno , Animales , Bacterias , Retroalimentación , Sistema Inmunológico
8.
PLoS Pathog ; 14(2): e1006847, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29394281

RESUMEN

Host responses to infection encompass many processes in addition to activation of the immune system, including metabolic adaptations, stress responses, tissue repair, and other reactions. The response to bacterial infection in Drosophila melanogaster has been classically described in studies that focused on the immune response elicited by a small set of largely avirulent microbes. Thus, we have surprisingly limited knowledge of responses to infection that are outside the canonical immune response, of how the response to pathogenic infection differs from that to avirulent bacteria, or even of how generic the response to various microbes is and what regulates that core response. In this study, we addressed these questions by profiling the D. melanogaster transcriptomic response to 10 bacteria that span the spectrum of virulence. We found that each bacterium triggers a unique transcriptional response, with distinct genes making up to one third of the response elicited by highly virulent bacteria. We also identified a core set of 252 genes that are differentially expressed in response to the majority of bacteria tested. Among these, we determined that the transcription factor CrebA is a novel regulator of infection tolerance. Knock-down of CrebA significantly increased mortality from microbial infection without any concomitant change in bacterial number. Upon infection, CrebA is upregulated by both the Toll and Imd pathways in the fat body, where it is required to induce the expression of secretory pathway genes. Loss of CrebA during infection triggered endoplasmic reticulum (ER) stress and activated the unfolded protein response (UPR), which contributed to infection-induced mortality. Altogether, our study reveals essential features of the response to bacterial infection and elucidates the function of a novel regulator of infection tolerance.


Asunto(s)
Proteína de Unión al Elemento de Respuesta al AMP Cíclico/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/inmunología , Drosophila melanogaster/metabolismo , Regulación del Desarrollo de la Expresión Génica , Interacciones Huésped-Patógeno , Tolerancia Inmunológica , Inmunidad Innata , Inmunidad Adaptativa , Animales , Animales Modificados Genéticamente , Carga Bacteriana , Vacunas Bacterianas/administración & dosificación , Proteína de Unión al Elemento de Respuesta al AMP Cíclico/antagonistas & inhibidores , Proteína de Unión al Elemento de Respuesta al AMP Cíclico/genética , Proteínas de Drosophila/antagonistas & inhibidores , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/microbiología , Estrés del Retículo Endoplásmico , Cuerpo Adiposo/inmunología , Cuerpo Adiposo/metabolismo , Cuerpo Adiposo/microbiología , Cuerpo Adiposo/patología , Perfilación de la Expresión Génica , Biblioteca de Genes , Bacterias Gramnegativas/crecimiento & desarrollo , Bacterias Gramnegativas/inmunología , Bacterias Gramnegativas/patogenicidad , Bacterias Gramnegativas/fisiología , Bacterias Grampositivas/crecimiento & desarrollo , Bacterias Grampositivas/inmunología , Bacterias Grampositivas/patogenicidad , Bacterias Grampositivas/fisiología , Masculino , Interferencia de ARN , Análisis de Supervivencia , Vacunas de Productos Inactivados/administración & dosificación , Virulencia
9.
Mol Biol Evol ; 34(4): 857-872, 2017 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-28087775

RESUMEN

The house fly, Musca domestica, occupies an unusual diversity of potentially septic niches compared with other sequenced Dipteran insects and is a vector of numerous diseases of humans and livestock. In the present study, we apply whole-transcriptome sequencing to identify genes whose expression is regulated in adult flies upon bacterial infection. We then combine the transcriptomic data with analysis of rates of gene duplication and loss to provide insight into the evolutionary dynamics of immune-related genes. Genes up-regulated after bacterial infection are biased toward being evolutionarily recent innovations, suggesting the recruitment of novel immune components in the M. domestica or ancestral Dipteran lineages. In addition, using new models of gene family evolution, we show that several different classes of immune-related genes, particularly those involved in either pathogen recognition or pathogen killing, are duplicating at a significantly accelerated rate on the M. domestica lineage relative to other Dipterans. Taken together, these results suggest that the M. domestica immune response includes an elevated diversity of genes, perhaps as a consequence of its lifestyle in septic environments.


Asunto(s)
Inmunidad Adaptativa/genética , Moscas Domésticas/genética , Animales , Secuencia de Bases/genética , Eliminación de Gen , Duplicación de Gen/genética , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/inmunología , Variación Genética/genética , Anotación de Secuencia Molecular , Tasa de Mutación , Transcriptoma/genética
10.
Proc Biol Sci ; 285(1870)2018 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-29321302

RESUMEN

Understanding the rate of evolutionary change and the genetic architecture that facilitates rapid adaptation is a current challenge in evolutionary biology. Comparative studies show that genes with immune function are among the most rapidly evolving genes across a range of taxa. Here, we use immune defence in natural populations of Drosophila melanogaster to understand the rate of evolution in natural populations and the genetics underlying rapid change. We probed the immune system using the natural pathogens Enterococcus faecalis and Providencia rettgeri to measure post-infection survival and bacterial load of wild D. melanogaster populations collected across seasonal time along a latitudinal transect along eastern North America (Massachusetts, Pennsylvania and Virginia). There are pronounced and repeatable changes in the immune response over the approximately 10 generations between spring and autumn collections, with a significant but less distinct difference observed among geographical locations. Genes with known immune function are not enriched among alleles that cycle with seasonal time, but the immune function of a subset of seasonally cycling alleles in immune genes was tested using reconstructed outbred populations. We find that flies containing seasonal alleles in Thioester-containing protein 3 (Tep3) have different functional responses to infection and that epistatic interactions among seasonal Tep3 and Drosomycin-like 6 (Dro6) alleles underlie the immune phenotypes observed in natural populations. This rapid, cyclic response to seasonal environmental pressure broadens our understanding of the complex ecological and genetic interactions determining the evolution of immune defence in natural populations.


Asunto(s)
Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Evolución Molecular , Inmunidad Innata/genética , Estaciones del Año , Adaptación Fisiológica , Animales , Proteínas de Drosophila/inmunología , Drosophila melanogaster/inmunología , Drosophila melanogaster/microbiología , Enterococcus faecalis , Femenino , Masculino , Massachusetts , Pennsylvania , Providencia , Virginia
11.
PLoS Biol ; 13(4): e1002136, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25915419

RESUMEN

Life history theory predicts that trait evolution should be constrained by competing physiological demands on an organism. Immune defense provides a classic example in which immune responses are presumed to be costly and therefore come at the expense of other traits related to fitness. One strategy for mitigating the costs of expensive traits is to render them inducible, such that the cost is paid only when the trait is utilized. In the current issue of PLOS Biology, Bajgar and colleagues elegantly demonstrate the energetic and life history cost of the immune response that Drosophila melanogaster larvae induce after infection by the parasitoid wasp Leptopilina boulardi. These authors show that infection-induced proliferation of defensive blood cells commands a diversion of dietary carbon away from somatic growth and development, with simple sugars instead being shunted to the hematopoetic organ for rapid conversion into the raw energy required for cell proliferation. This metabolic shift results in a 15% delay in the development of the infected larva and is mediated by adenosine signaling between the hematopoietic organ and the central metabolic control organ of the host fly. The adenosine signal thus allows D. melanogaster to rapidly marshal the energy needed for effective defense and to pay the cost of immunity only when infected.


Asunto(s)
Adenosina/fisiología , Drosophila/inmunología , Sistema Inmunológico/fisiología , Avispas/fisiología , Animales
12.
PLoS Biol ; 13(7): e1002210, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26172158

RESUMEN

Drosophila melanogaster is a valuable invertebrate model for viral infection and antiviral immunity, and is a focus for studies of insect-virus coevolution. Here we use a metagenomic approach to identify more than 20 previously undetected RNA viruses and a DNA virus associated with wild D. melanogaster. These viruses not only include distant relatives of known insect pathogens but also novel groups of insect-infecting viruses. By sequencing virus-derived small RNAs, we show that the viruses represent active infections of Drosophila. We find that the RNA viruses differ in the number and properties of their small RNAs, and we detect both siRNAs and a novel miRNA from the DNA virus. Analysis of small RNAs also allows us to identify putative viral sequences that lack detectable sequence similarity to known viruses. By surveying >2,000 individually collected wild adult Drosophila we show that more than 30% of D. melanogaster carry a detectable virus, and more than 6% carry multiple viruses. However, despite a high prevalence of the Wolbachia endosymbiont--which is known to be protective against virus infections in Drosophila--we were unable to detect any relationship between the presence of Wolbachia and the presence of any virus. Using publicly available RNA-seq datasets, we show that the community of viruses in Drosophila laboratories is very different from that seen in the wild, but that some of the newly discovered viruses are nevertheless widespread in laboratory lines and are ubiquitous in cell culture. By sequencing viruses from individual wild-collected flies we show that some viruses are shared between D. melanogaster and D. simulans. Our results provide an essential evolutionary and ecological context for host-virus interaction in Drosophila, and the newly reported viral sequences will help develop D. melanogaster further as a model for molecular and evolutionary virus research.


Asunto(s)
Evolución Biológica , Drosophila melanogaster/virología , Secuencia de Aminoácidos , Animales , Secuencia Conservada , Drosophila simulans/virología , Femenino , Masculino , Metagenómica , Datos de Secuencia Molecular , ARN/análisis , Interferencia de ARN , Proteínas Virales/química , Wolbachia/aislamiento & purificación
13.
Biol Lett ; 14(2)2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29438055

RESUMEN

Organisms with complex life cycles can differ markedly in their biology across developmental life stages. Consequently, distinct life stages can represent drastically different environments for parasites. This difference is especially striking with holometabolous insects, which have dramatically different larval and adult life stages, bridged by a complete metamorphosis. There is no a priori guarantee that a parasite infecting the larval stage would be able to persist into the adult stage. In fact, to our knowledge, transstadial transmission of extracellular pathogens has never been documented in a host that undergoes complete metamorphosis. We tested the hypothesis that a bacterial parasite originally sampled from an adult host could infect a larva, then survive through metamorphosis and persist into the adult stage. As a model, we infected the host Drosophila melanogaster with a horizontally transmitted, extracellular bacterial pathogen, Providencia rettgeri We found that this natural pathogen survived systemic infection of larvae (L3) and successfully persisted into the adult host. We then discuss how it may be adaptive for bacteria to transverse life stages and even minimize virulence at the larval stage in order to benefit from adult dispersal.


Asunto(s)
Drosophila melanogaster/microbiología , Metamorfosis Biológica , Providencia/fisiología , Animales , Larva/microbiología
14.
PLoS Genet ; 11(3): e1005030, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25764027

RESUMEN

Both malnutrition and undernutrition can lead to compromised immune defense in a diversity of animals, and "nutritional immunology" has been suggested as a means of understanding immunity and determining strategies for fighting infection. The genetic basis for the effects of diet on immunity, however, has been largely unknown. In the present study, we have conducted genome-wide association mapping in Drosophila melanogaster to identify the genetic basis for individual variation in resistance, and for variation in immunological sensitivity to diet (genotype-by-environment interaction, or GxE). D. melanogaster were reared for several generations on either high-glucose or low-glucose diets and then infected with Providencia rettgeri, a natural bacterial pathogen of D. melanogaster. Systemic pathogen load was measured at the peak of infection intensity, and several indicators of nutritional status were taken from uninfected flies reared on each diet. We find that dietary glucose level significantly alters the quality of immune defense, with elevated dietary glucose resulting in higher pathogen loads. The quality of immune defense is genetically variable within the sampled population, and we find genetic variation for immunological sensitivity to dietary glucose (genotype-by-diet interaction). Immune defense was genetically correlated with indicators of metabolic status in flies reared on the high-glucose diet, and we identified multiple genes that explain variation in immune defense, including several that have not been previously implicated in immune response but which are confirmed to alter pathogen load after RNAi knockdown. Our findings emphasize the importance of dietary composition to immune defense and reveal genes outside the conventional "immune system" that can be important in determining susceptibility to infection. Functional variation in these genes is segregating in a natural population, providing the substrate for evolutionary response to pathogen pressure in the context of nutritional environment.


Asunto(s)
Dieta , Interacción Gen-Ambiente , Interacciones Huésped-Patógeno/genética , Inmunidad Innata/genética , Animales , Drosophila melanogaster , Variación Genética , Estudio de Asociación del Genoma Completo , Glucosa/metabolismo , Interacciones Huésped-Patógeno/inmunología , Providencia/inmunología , Providencia/patogenicidad
15.
BMC Biol ; 15(1): 124, 2017 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-29268741

RESUMEN

BACKGROUND: Host sexual dimorphism is being increasingly recognized to generate strong differences in the outcome of infectious disease, but the mechanisms underlying immunological differences between males and females remain poorly characterized. Here, we used Drosophila melanogaster to assess and dissect sexual dimorphism in the innate response to systemic bacterial infection. RESULTS: We demonstrated sexual dimorphism in susceptibility to infection by a broad spectrum of Gram-positive and Gram-negative bacteria. We found that both virgin and mated females are more susceptible than mated males to most, but not all, infections. We investigated in more detail the lower resistance of females to infection with Providencia rettgeri, a Gram-negative bacterium that naturally infects D. melanogaster. We found that females have a higher number of phagocytes than males and that ablation of hemocytes does not eliminate the dimorphism in resistance to P. rettgeri, so the observed dimorphism does not stem from differences in the cellular response. The Imd pathway is critical for the production of antimicrobial peptides in response to Gram-negative bacteria, but mutants for Imd signaling continued to exhibit dimorphism even though both sexes showed strongly reduced resistance. Instead, we found that the Toll pathway is responsible for the dimorphism in resistance. The Toll pathway is dimorphic in genome-wide constitutive gene expression and in induced response to infection. Toll signaling is dimorphic in both constitutive signaling and in induced activation in response to P. rettgeri infection. The dimorphism in pathway activation can be specifically attributed to Persephone-mediated immune stimulation, by which the Toll pathway is triggered in response to pathogen-derived virulence factors. We additionally found that, in absence of Toll signaling, males become more susceptible than females to the Gram-positive Enterococcus faecalis. This reversal in susceptibility between male and female Toll pathway mutants compared to wildtype hosts highlights the key role of the Toll pathway in D. melanogaster sexual dimorphism in resistance to infection. CONCLUSION: Altogether, our data demonstrate that Toll pathway activity differs between male and female D. melanogaster in response to bacterial infection, thus identifying innate immune signaling as a determinant of sexual immune dimorphism.


Asunto(s)
Proteínas de Drosophila/genética , Proteínas de Drosophila/inmunología , Drosophila melanogaster/genética , Drosophila melanogaster/microbiología , Bacterias Gramnegativas/fisiología , Bacterias Grampositivas/fisiología , Receptores Toll-Like/inmunología , Animales , Resistencia a la Enfermedad/genética , Drosophila melanogaster/inmunología , Femenino , Bacterias Gramnegativas/inmunología , Bacterias Grampositivas/inmunología , Masculino , Caracteres Sexuales
16.
Proc Biol Sci ; 284(1869)2017 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-29263278

RESUMEN

The role of microbial secondary metabolites in the ecology of the organisms that produce them remains poorly understood. Variation in aflatoxin production by Aspergillus flavus is maintained by balancing selection, but the ecological function and impact on fungal fitness of this compound are unknown. We hypothesize that balancing selection for aflatoxin production in A. flavus is driven by interaction with insects. To test this, we competed naturally occurring aflatoxigenic and non-aflatoxigenic fungal isolates against Drosophila larvae on medium containing 0-1750 ppb aflatoxin, using quantitative PCR to quantify A. flavus DNA as a proxy for fungal fitness. The addition of aflatoxin across this range resulted in a 26-fold increase in fungal fitness. With no added toxin, aflatoxigenic isolates caused higher mortality of Drosophila larvae and had slightly higher fitness than non-aflatoxigenic isolates. Additionally, aflatoxin production increased an average of 1.5-fold in the presence of a single larva and nearly threefold when the fungus was mechanically damaged. We argue that the role of aflatoxin in protection from fungivory is inextricably linked to its role in interference competition. Our results, to our knowledge, provide the first clear evidence of a fitness advantage conferred to A. flavus by aflatoxin when interacting with insects.


Asunto(s)
Aflatoxinas/metabolismo , Aspergillus flavus/química , Aspergillus flavus/genética , Drosophila melanogaster/fisiología , Herbivoria , Selección Genética , Animales , Drosophila melanogaster/crecimiento & desarrollo , Microbiología de Alimentos , Larva/crecimiento & desarrollo , Larva/fisiología
17.
Mol Ecol ; 26(6): 1533-1546, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28099780

RESUMEN

Defence against pathogenic infection can take two forms: resistance and tolerance. Resistance is the ability of the host to limit a pathogen burden, whereas tolerance is the ability to limit the negative consequences of infection at a given level of infection intensity. Evolutionarily, a tolerance strategy that is independent of resistance could allow the host to avoid mounting a costly immune response and, theoretically, to avoid a co-evolutionary arms race between pathogen virulence and host resistance. Biomedically, understanding the mechanisms of tolerance and how they relate to resistance could potentially yield treatment strategies that focus on health improvement instead of pathogen elimination. To understand the impact of tolerance on host defence and identify genetic variants that determine host tolerance, we defined genetic variation in tolerance as the residual deviation from a binomial regression of fitness under infection against infection intensity. We then performed a genomewide association study to map the genetic basis of variation in resistance to and tolerance of infection by the bacterium Providencia rettgeri. We found a positive genetic correlation between resistance and tolerance, and we demonstrated that the level of resistance is highly predictive of tolerance. We identified 30 loci that predict tolerance, many of which are in genes involved in the regulation of immunity and metabolism. We used RNAi to confirm that a subset of mapped genes have a role in defence, including putative wound repair genes grainy head and debris buster. Our results indicate that tolerance is not an independent strategy from resistance, but that defence arises from a collection of physiological processes intertwined with canonical immunity and resistance.


Asunto(s)
Infecciones Bacterianas/genética , Resistencia a la Enfermedad/genética , Drosophila melanogaster/genética , Drosophila melanogaster/microbiología , Variación Genética , Animales , Evolución Biológica , Genes de Insecto , Aptitud Genética , Estudio de Asociación del Genoma Completo
18.
Annu Rev Entomol ; 61: 239-56, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26667271

RESUMEN

Immune defense and reproduction are physiologically and energetically demanding processes and have been observed to trade off in a diversity of female insects. Increased reproductive effort results in reduced immunity, and reciprocally, infection and activation of the immune system reduce reproductive output. This trade-off can manifest at the physiological level (within an individual) and at the evolutionary level (genetic distinction among individuals in a population). The resource allocation model posits that the trade-off arises because of competition for one or more limiting resources, and we hypothesize that pleiotropic signaling mechanisms regulate allocation of that resource between reproductive and immune processes. We examine the role of juvenile hormone, 20-hydroxyecdysone, and insulin/insulin-like growth factor-like signaling in regulating both oogenesis and immune system activity, and propose a signaling network that may mechanistically regulate the trade-off. Finally, we discuss implications of the trade-off in an ecological and evolutionary context.


Asunto(s)
Sistema Inmunológico , Insectos/fisiología , Oogénesis , Transducción de Señal , Animales , Hormonas de Insectos/metabolismo , Insectos/inmunología , Insulina/metabolismo , Somatomedinas/metabolismo
19.
Mol Ecol ; 25(7): 1494-510, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26846876

RESUMEN

The recent discovery of a previously unknown genetic subgroup of Anopheles gambiae sensu lato underscores our incomplete understanding of complexities of vector population demographics in Anopheles. This subgroup, named GOUNDRY, does not rest indoors as adults and is highly susceptible to Plasmodium infection in the laboratory. Initial description of GOUNDRY suggested it differed from other known Anopheles taxa in surprising and sometimes contradictory ways, raising a number of questions about its age, population size and relationship to known subgroups. To address these questions, we sequenced the complete genomes of 12 wild-caught GOUNDRY specimens and compared these genomes to a panel of Anopheles genomes. We show that GOUNDRY is most closely related to Anopheles coluzzii, and the timing of cladogenesis is not recent, substantially predating the advent of agriculture. We find a large region of the X chromosome that has swept to fixation in GOUNDRY within the last 100 years, which may be an inversion that serves as a partial barrier to contemporary gene flow. Interestingly, we show that GOUNDRY has a history of inbreeding that is significantly associated with susceptibility to Plasmodium infection in the laboratory. Our results illuminate the genomic evolution of one of probably several cryptic, ecologically specialized subgroups of Anopheles and provide a potent example of how vector population dynamics may complicate efforts to control or eradicate malaria.


Asunto(s)
Anopheles/genética , Evolución Molecular , Genoma de los Insectos , Plasmodium falciparum , Animales , Anopheles/parasitología , Inversión Cromosómica , Flujo Génico , Especiación Genética , Genética de Población , Endogamia , Insectos Vectores/genética , Insectos Vectores/parasitología , Polimorfismo de Nucleótido Simple , Dinámica Poblacional , Análisis de Secuencia de ADN , Cromosoma X/genética
20.
Infect Immun ; 82(10): 4380-9, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25092914

RESUMEN

The route of infection can profoundly affect both the progression and outcome of disease. We investigated differences in Drosophila melanogaster defense against infection after bacterial inoculation into two sites--the abdomen and the thorax. Thorax inoculation results in increased bacterial proliferation and causes high mortality within the first few days of infection. In contrast, abdomen inoculation results in minimal mortality and lower bacterial loads than thorax inoculation. Inoculation into either site causes systemic infection. Differences in mortality and bacterial load are due to injury of the thorax and can be recapitulated by abdominal inoculation coupled with aseptic wounding of the thorax. This altered resistance appears to be independent of classical immune pathways and opens new avenues of research on the role of injury during defense against infection.


Asunto(s)
Drosophila melanogaster/fisiología , Traumatismos Abdominales/inmunología , Animales , Infecciones Bacterianas/inmunología , Infecciones Bacterianas/microbiología , Carga Bacteriana , Modelos Animales de Enfermedad , Resistencia a la Enfermedad , Drosophila melanogaster/inmunología , Femenino , Masculino , Análisis de Supervivencia , Traumatismos Torácicos/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA