Abstract

Intracellular bacterial pathogens remodel cellular functions during their infectious cycle via the coordinated actions of effector molecules delivered through dedicated secretion systems. While the function of many individual effectors is known, how they interact to promote pathogenesis is rarely understood. The zoonotic bacterium Brucella abortus, the causative agent of brucellosis, delivers effector proteins via its VirB type IV secretion system (T4SS) which mediate biogenesis of the endoplasmic reticulum (ER)-derived replicative Brucella-containing vacuole (rBCV). Here, we show that T4SS effectors BspB and RicA display epistatic interactions in Brucella replication. Defects in rBCV biogenesis and Brucella replication caused by deletion of bspB were dependent on the host GTPase Rab2a and suppressed by the deletion of ricA, indicating a role of Rab2-binding effector RicA in these phenotypic defects. Rab2a requirements for rBCV biogenesis and Brucella intracellular replication were abolished upon deletion of both bspB and ricA, demonstrating that the functional interaction of these effectors engages Rab2-dependent transport in the Brucella intracellular cycle. Expression of RicA impaired host secretion and caused Golgi fragmentation. While BspB-mediated changes in ER-to-Golgi transport were independent of RicA and Rab2a, BspB-driven alterations in Golgi vesicular traffic also involved RicA and Rab2a, defining BspB and RicA's functional interplay at the Golgi interface. Altogether, these findings support a model where RicA modulation of Rab2a functions impairs Brucella replication but is compensated by BspB-mediated remodeling of Golgi apparatus-associated vesicular transport, revealing an epistatic interaction between these T4SS effectors.IMPORTANCE Bacterial pathogens with an intracellular lifestyle modulate many host cellular processes to promote their infectious cycle. They do so by delivering effector proteins into host cells via dedicated secretion systems that target specific host functions. While the roles of many individual effectors are known, how their modes of action are coordinated is rarely understood. Here, we show that the zoonotic bacterium Brucella abortus delivers the BspB effector that mitigates the negative effect on bacterial replication that the RicA effector exerts via modulation of the host small GTPase Rab2. These findings provide an example of functional integration between bacterial effectors that promotes proliferation of pathogens.

Highlights

  • Intracellular bacterial pathogens remodel cellular functions during their infectious cycle via the coordinated actions of effector molecules delivered through dedicated secretion systems

  • The deletion of ricA in ΔbspB bacteria suppressed the delayed replicative Brucella-containing vacuole (rBCV) biogenesis and bacterial replication defect caused by deletion of bspB seen in bone marrowderived macrophages (BMMs), indicating that BspB is dispensable for these intracellular events in the absence of RicA

  • We show that BspB and RicA, two effectors previously associated with rBCV biogenesis and bacterial replication (14, 17, 22), comodulate Golgi apparatusassociated vesicular transport and engage Rab2-dependent functions in Brucella intracellular replication

Read more

Summary

Introduction

Intracellular bacterial pathogens remodel cellular functions during their infectious cycle via the coordinated actions of effector molecules delivered through dedicated secretion systems. Intracellular bacterial pathogens have developed an array of mechanisms to exploit and redirect host cellular functions toward achieving their infectious cycle For this purpose, many use dedicated secretion systems that deliver effector proteins into host cells and which individually target and modulate specific host functions to promote bacterial survival, persistence, or proliferation. BspB alters secretory traffic via its interaction with the Golgi apparatus-associated conserved oligomeric Golgi (COG) complex, a regulator of vesicular traffic at, and within, the Golgi apparatus (23) and causes redirection of COG-dependent vesicular traffic to promote rBCV biogenesis and bacterial replication (22) These findings argue that Brucella delivers a series of effector proteins that modulate specific host secretory functions to mediate rBCV biogenesis and possibly bacterial replication within rBCVs. Whether, and how, Brucella T4SS effectors coordinate to integrate their respective modes of action toward promoting the bacterium’s intracellular cycle is unknown

Methods
Results
Conclusion
Full Text
Paper version not known

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

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.