Abstract

BackgroundHost 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.ResultsWe 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.ConclusionAltogether, 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.

Highlights

  • 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

  • The Imd pathway is activated upon the detection of peptidoglycan produced by Gram-negative bacteria, whereas the Toll pathway responds to the peptidoglycan of most Gram-positive bacteria and to proteases secreted during pathogenic infections [13]

  • Sexual dimorphism in host susceptibility to infection is pervasive In order to test whether D. melanogaster are sexually dimorphic in survivorship in response to infection, we first injected a genetically diverse population as well as several inbred populations of D. melanogaster with both

Read more

Summary

Introduction

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. We used Drosophila melanogaster to assess and dissect sexual dimorphism in the innate response to systemic bacterial infection. The most striking differences among individuals in a population are often those between the sexes Such dimorphism is often characterized by obvious differences in morphology and behavior, as well as in a number of differences in physiological functions, including immunity, metabolism, and disease outcome [1]. Despite the plethora of examples of sexual dimorphism in disease outcomes [3, 5, 6], the characterization of these differences have been largely overlooked in medical studies and in studies of natural systems [7,8,9,10,11]. We used Drosophila melanogaster to understand the basis of sexual dimorphism of an innate immune system.

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call