Abstract

Interferon-regulated immune defenses protect mammals from pathogenically diverse obligate intracellular bacterial pathogens of the genus Chlamydia Interferon gamma (IFN-γ) is especially important in controlling the virulence of Chlamydia species and thus impacts the modeling of human chlamydial infection and disease in mice. How IFN-γ contributes to cell-autonomous defenses against Chlamydia species and how these pathogens evade IFN-γ-mediated immunity in their natural hosts are not well understood. We conducted a genetic screen which identified 31 IFN-γ-sensitive (Igs) mutants of the mouse model pathogen Chlamydia muridarum Genetic suppressor analysis and lateral gene transfer were used to map the phenotype of one of these mutants, Igs4, to a missense mutation in a putative chlamydial inclusion membrane protein, TC0574. We observed the lytic destruction of Igs4-occupied inclusions and accompanying host cell death in response to IFN-γ priming or various proapoptotic stimuli. However, Igs4 was insensitive to IFN-γ-regulated cell-autonomous defenses previously implicated in anti-Chlamydia trachomatis host defense in mice. Igs4 inclusion integrity was restored by caspase inhibitors, indicating that the IFN-γ-mediated destruction of Igs4 inclusions is dependent upon the function of caspases or related prodeath cysteine proteases. We further demonstrated that the Igs4 mutant is immune restricted in an IFN-γ-dependent manner in a mouse infection model, thereby implicating IFN-γ-mediated inclusion destruction and host cell death as potent in vivo host defense mechanisms to which wild-type C. muridarum is resistant. Overall, our results suggest that C. muridarum evolved resistance mechanisms to counter IFN-γ-elicited programmed cell death and the associated destruction of intravacuolar pathogens.IMPORTANCE Multiple obligatory intracellular bacteria in the genus Chlamydia are important pathogens. In humans, strains of C. trachomatis cause trachoma, chlamydia, and lymphogranuloma venereum. These diseases are all associated with extended courses of infection and reinfection that likely reflect the ability of chlamydiae to evade various aspects of host immune responses. Interferon-stimulated genes, driven in part by the cytokine interferon gamma, restrict the host range of various Chlamydia species, but how these pathogens evade interferon-stimulated genes in their definitive host is poorly understood. Various Chlamydia species can inhibit death of their host cells and may have evolved this strategy to evade prodeath signals elicited by host immune responses. We present evidence that chlamydia-induced programmed cell death resistance evolved to counter interferon- and immune-mediated killing of Chlamydia-infected cells.

Highlights

  • IMPORTANCE Multiple obligatory intracellular bacteria in the genus Chlamydia are important pathogens

  • Our results show that chlamydial programmed cell death resistance (PCDR) and interferon resistance are linked and suggest that C. muridarum has evolved mechanisms to protect inclusions against an IFN-␥-triggered membranolytic pathway executed by prodeath cysteine proteases

  • We identified 31 IFN-␥-sensitive (Igs) C. muridarum mutants that were more sensitive to IFN-␥ than wild-type C. muridarum

Read more

Summary

Introduction

IMPORTANCE Multiple obligatory intracellular bacteria in the genus Chlamydia are important pathogens. C. trachomatis and Chlamydia muridarum can block cell death in epithelial cells exposed to prodeath signals [7, 8] These observations suggest that Chlamydia spp. counter intracellular immune defenses while maintaining the viability of their host cell to complete a productive developmental cycle. In contrast to C. trachomatis, mice are significantly more susceptible to infection and disease mediated by the rodent-adapted pathogen C. muridarum, presumably because C. muridarum encodes host-tailored virulence factors that help it counter the relevant IFN-␥-regulated cell-autonomous defenses of its definitive murine host [5, 16]. Infection with Inc mutants deficient for inclusion maintenance triggers host cell death dependent on the autophagy protein Beclin and the cytosolic immune sensor stimulator of interferon genes (STING) but independent of the STING-mediated type I IFN response [24]. Chlamydia Interferon and Cell Death Evasion Are Linked response leading to inclusion disruption nor the chlamydial factors counteracting this membranolytic host defense program have been identified

Objectives
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