Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters











Database
Language
Publication year range
1.
PLoS Pathog ; 18(9): e1010826, 2022 09.
Article in English | MEDLINE | ID: mdl-36129961

ABSTRACT

Host behavioural changes are among the most apparent effects of infection. 'Sickness behaviour' can involve a variety of symptoms, including anorexia, depression, and changed activity levels. Here, using a real-time tracking and behavioural profiling platform, we show that in Drosophila melanogaster, several systemic bacterial infections cause significant increases in physical activity, and that the extent of this activity increase is a predictor of survival time in some lethal infections. Using multiple bacteria and D. melanogaster immune and activity mutants, we show that increased activity is driven by at least two different mechanisms. Increased activity after infection with Micrococcus luteus, a Gram-positive bacterium rapidly cleared by the immune response, strictly requires the Toll ligand spätzle. In contrast, increased activity after infection with Francisella novicida, a Gram-negative bacterium that cannot be cleared by the immune response, is entirely independent of both Toll and the parallel IMD pathway. The existence of multiple signalling mechanisms by which bacterial infections drive increases in physical activity implies that this effect may be an important aspect of the host response.


Subject(s)
Drosophila Proteins , Drosophila melanogaster , Animals , Drosophila Proteins/metabolism , Drosophila melanogaster/microbiology , Gram-Negative Bacteria , Gram-Positive Bacteria , Immunity, Innate , Ligands
2.
Proc Natl Acad Sci U S A ; 118(32)2021 08 10.
Article in English | MEDLINE | ID: mdl-34341118

ABSTRACT

Male and female animals exhibit differences in infection outcomes. One possible source of sexually dimorphic immunity is the sex-specific costs of immune activity or pathology, but little is known about the independent effects of immune- versus microbe-induced pathology and whether these may differ for the sexes. Here, by measuring metabolic and physiological outputs in Drosophila melanogaster with wild-type and mutant immune responses, we test whether the sexes are differentially impacted by these various sources of pathology and identify a critical regulator of this difference. We find that the sexes exhibit differential immune activity but similar bacteria-derived metabolic pathology. We show that female-specific immune-inducible expression of PGRP-LB, a negative regulator of the immune deficiency (IMD) pathway, enables females to reduce immune activity in response to reductions in bacterial numbers. In the absence of PGRP-LB, females are more resistant to infection, confirming the functional importance of this regulation and suggesting that female-biased immune restriction comes at a cost.


Subject(s)
Carrier Proteins/immunology , Drosophila melanogaster/immunology , Drosophila melanogaster/microbiology , Animals , Carrier Proteins/genetics , Drosophila Proteins/genetics , Drosophila Proteins/immunology , Drosophila melanogaster/genetics , Escherichia coli Infections/immunology , Escherichia coli Infections/veterinary , Female , Gene Expression Regulation , Immunity, Innate/physiology , Male , Mutation , Nuclear Proteins/genetics , Nuclear Proteins/immunology , Repressor Proteins/genetics , Repressor Proteins/immunology , Sex Factors , Signal Transduction/physiology , Triglycerides/metabolism
3.
Front Immunol ; 11: 1419, 2020.
Article in English | MEDLINE | ID: mdl-32733472

ABSTRACT

The origins and causes of infection pathologies are often not understood. Despite this, the study of infection and immunity relies heavily on the ability to discern between potential sources of pathology. Work in the fruit fly has supported the assumption that mortality resulting from bacterial invasion is largely due to direct host-pathogen interactions, as lower pathogen loads are often associated with reduced pathology, and bacterial load upon death is predictable. However, the mechanisms through which these interactions bring about host death are complex. Here we show that infection with the bacterium Francisella novicida leads to metabolic dysregulation and, using treatment with a bacteriostatic antibiotic, we show that this pathology is the result of direct interaction between host and pathogen. We show that mutants of the immune deficiency immune pathway fail to exhibit similar metabolic dysregulation, supporting the idea that the reallocation of resources for immune-related activities contributes to metabolic dysregulation. Targeted investigation into the cross-talk between immune and metabolic pathways has the potential to illuminate some of this interaction.


Subject(s)
Bacterial Load/immunology , Gram-Negative Bacterial Infections/immunology , Gram-Negative Bacterial Infections/metabolism , Host-Pathogen Interactions/immunology , Animals , Anti-Bacterial Agents/pharmacology , Bacterial Load/drug effects , Drosophila melanogaster , Francisella , Gram-Negative Bacterial Infections/microbiology , Host-Pathogen Interactions/drug effects , Tetracycline/pharmacology
4.
Elife ; 92020 01 20.
Article in English | MEDLINE | ID: mdl-31944178

ABSTRACT

Unpaired ligands are secreted signals that act via a GP130-like receptor, domeless, to activate JAK/STAT signalling in Drosophila. Like many mammalian cytokines, unpaireds can be activated by infection and other stresses and can promote insulin resistance in target tissues. However, the importance of this effect in non-inflammatory physiology is unknown. Here, we identify a requirement for unpaired-JAK signalling as a metabolic regulator in healthy adult Drosophila muscle. Adult muscles show basal JAK-STAT signalling activity in the absence of any immune challenge. Plasmatocytes (Drosophila macrophages) are an important source of this tonic signal. Loss of the dome receptor on adult muscles significantly reduces lifespan and causes local and systemic metabolic pathology. These pathologies result from hyperactivation of AKT and consequent deregulation of metabolism. Thus, we identify a cytokine signal that must be received in muscle to control AKT activity and metabolic homeostasis.


Subject(s)
Cytokines/metabolism , Drosophila Proteins , Muscles/metabolism , Proto-Oncogene Proteins c-akt , Receptors, Interleukin , Signal Transduction/genetics , Animals , Animals, Genetically Modified , Drosophila/genetics , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Homeostasis , Janus Kinases/genetics , Janus Kinases/metabolism , Male , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Interleukin/genetics , Receptors, Interleukin/metabolism , STAT Transcription Factors/genetics , STAT Transcription Factors/metabolism
5.
Proc Biol Sci ; 281(1790)2014 09 07.
Article in English | MEDLINE | ID: mdl-25030981

ABSTRACT

Sex differences in immunity are often observed, with males generally having a weaker immune system than females. However, recent data in a sex-role-reversed species in which females compete to mate with males suggest that sexually competitive females have a weaker immune response. These findings support the hypothesis that sexual dimorphism in immunity has evolved in response to sex-specific fitness returns of investment in traits such as parental investment and longevity, but the scarcity of data in sex-reversed species prevents us from drawing general conclusions. Using an insect species in which males make a large but variable parental investment in their offspring, we use two indicators of immunocompetence to test the hypothesis that sex-biased immunity is determined by differences in parental investment. We found that when the value of paternal investment was experimentally increased, male immune investment became relatively greater than that of females. Thus, in this system, in which the direction of sexual competition is plastic, the direction of sex-biased immunity is also plastic and appears to track relative parental investment.


Subject(s)
Immunocompetence/physiology , Orthoptera/immunology , Reproduction/physiology , Sex Characteristics , Adaptation, Physiological/immunology , Animal Nutritional Physiological Phenomena , Animals , Female , Male , Monophenol Monooxygenase/immunology , Orthoptera/physiology , Phenotype , Sexual Behavior, Animal/physiology
6.
Proc Biol Sci ; 281(1788): 20140987, 2014 Aug 07.
Article in English | MEDLINE | ID: mdl-24966317

ABSTRACT

A critical task in evolutionary genetics is to explain the persistence of heritable variation in fitness-related traits such as immunity. Ecological factors can maintain genetic variation in immunity, but less is known about the role of other factors, such as antagonistic pleiotropy, on immunity. Sexually dimorphic immunity-with females often being more immune-competent-may maintain variation in immunity in dioecious populations. Most eco-immunological studies assess host resistance to parasites rather than the host's ability to maintain fitness during infection (tolerance). Distinguishing between resistance and tolerance is important as they are thought to have markedly different evolutionary and epidemiological outcomes. Few studies have investigated tolerance in animals, and the extent of sexual dimorphism in tolerance is unknown. Using males and females from 50 Drosophila melanogaster genotypes, we investigated possible sources of genetic variation for immunity by assessing both resistance and tolerance to the common bacterial pathogen Pseudomonas aeruginosa. We found evidence of sexual dimorphism and sexual antagonism for resistance and tolerance, and a trade-off between the two traits. Our findings suggest that antagonistic pleiotropy may be a major contributor to variation in immunity, with implications for host-parasite coevolution.


Subject(s)
Drosophila melanogaster/genetics , Drosophila melanogaster/microbiology , Genetic Variation , Immune Tolerance , Pseudomonas aeruginosa/physiology , Animals , Drosophila melanogaster/immunology , Female , Genotype , Male , Mutation , Sex Characteristics
SELECTION OF CITATIONS
SEARCH DETAIL