Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
Add more filters










Publication year range
1.
J Clin Microbiol ; 61(8): e0043823, 2023 08 23.
Article in English | MEDLINE | ID: mdl-37395662

ABSTRACT

Bacteria of the genus Brucella are facultative intracellular parasites that cause brucellosis, a severe animal and human disease. Recently, a group of taxonomists merged the brucellae with the primarily free-living, phylogenetically related Ochrobactrum spp. in the genus Brucella. This change, founded only on global genomic analysis and the fortuitous isolation of some opportunistic Ochrobactrum spp. from medically compromised patients, has been automatically included in culture collections and databases. We argue that clinical and environmental microbiologists should not accept this nomenclature, and we advise against its use because (i) it was presented without in-depth phylogenetic analyses and did not consider alternative taxonomic solutions; (ii) it was launched without the input of experts in brucellosis or Ochrobactrum; (iii) it applies a non-consensus genus concept that disregards taxonomically relevant differences in structure, physiology, population structure, core-pangenome assemblies, genome structure, genomic traits, clinical features, treatment, prevention, diagnosis, genus description rules, and, above all, pathogenicity; and (iv) placing these two bacterial groups in the same genus creates risks for veterinarians, medical doctors, clinical laboratories, health authorities, and legislators who deal with brucellosis, a disease that is particularly relevant in low- and middle-income countries. Based on all this information, we urge microbiologists, bacterial collections, genomic databases, journals, and public health boards to keep the Brucella and Ochrobactrum genera separate to avoid further bewilderment and harm.


Subject(s)
Brucella , Ochrobactrum , Ochrobactrum/classification , Ochrobactrum/genetics , Ochrobactrum/pathogenicity , Ochrobactrum/physiology , Brucella/classification , Brucella/genetics , Brucella/pathogenicity , Brucella/physiology , Terminology as Topic , Phylogeny , Brucellosis/drug therapy , Brucellosis/microbiology , Humans , Opportunistic Infections/microbiology
2.
Cell Host Microbe ; 31(8): 1359-1370.e7, 2023 08 09.
Article in English | MEDLINE | ID: mdl-37453420

ABSTRACT

Glutathione (GSH) is an abundant metabolite within eukaryotic cells that can act as a signal, a nutrient source, or serve in a redox capacity for intracellular bacterial pathogens. For Francisella, GSH is thought to be a critical in vivo source of cysteine; however, the cellular pathways permitting GSH utilization by Francisella differ between strains and have remained poorly understood. Using genetic screening, we discovered a unique pathway for GSH utilization in Francisella. Whereas prior work suggested GSH catabolism initiates in the periplasm, the pathway we define consists of a major facilitator superfamily (MFS) member that transports intact GSH and a previously unrecognized bacterial cytoplasmic enzyme that catalyzes the first step of GSH degradation. Interestingly, we find that the transporter gene for this pathway is pseudogenized in pathogenic Francisella, explaining phenotypic discrepancies in GSH utilization among Francisella spp. and revealing a critical role for GSH in the environmental niche of these bacteria.


Subject(s)
Francisella tularensis , Francisella , Glutathione/metabolism , Francisella/genetics , Francisella/metabolism , Francisella tularensis/genetics , Francisella tularensis/growth & development , Francisella tularensis/metabolism , DNA Transposable Elements , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Phylogeny , Macrophages/parasitology , Animals , Mice , Tularemia/microbiology
3.
Infect Immun ; 91(5): e0013023, 2023 05 16.
Article in English | MEDLINE | ID: mdl-37129527

ABSTRACT

Brucella abortus, the intracellular causative agent of brucellosis, relies on type IV secretion system (T4SS) effector-mediated modulation of host cell functions to establish a replicative niche, the Brucella-containing vacuole (BCV). Brucella exploits the host's endocytic, secretory, and autophagic pathways to modulate the nature and function of its vacuole from an endocytic BCV (eBCV) to an endoplasmic reticulum (ER)-derived replicative BCV (rBCV) to an autophagic egress BCV (aBCV). A role for the host ER-associated degradation pathway (ERAD) in the B. abortus intracellular cycle was recently uncovered, as it is enhanced by the T4SS effector BspL to control the timing of aBCV-mediated egress. Here, we show that the T4SS effector BspA also interferes with ERAD, yet to promote B. abortus intracellular proliferation. BspA was required for B. abortus replication in bone marrow-derived macrophages and interacts with membrane-associated RING-CH-type finger 6 (MARCH6), a host E3 ubiquitin ligase involved in ERAD. Pharmacological inhibition of ERAD and small interfering RNA (siRNA) depletion of MARCH6 did not affect the replication of wild-type B. abortus but rescued the replication defect of a bspA deletion mutant, while depletion of the ERAD component UbxD8 affected replication of B. abortus and rescued the replication defect of the bspA mutant. BspA affected the degradation of ERAD substrates and destabilized the MARCH6 E3 ligase complex. Taken together, these findings indicate that BspA inhibits the host ERAD pathway via targeting of MARCH6 to promote B. abortus intracellular growth. Our data reveal that targeting ERAD components by type IV effectors emerges as a multifaceted theme in Brucella pathogenesis.


Subject(s)
Bacterial Proteins , Brucella abortus , Brucellosis , Membrane Proteins , Type IV Secretion Systems , Animals , Mice , Brucella abortus/physiology , Type IV Secretion Systems/metabolism , Brucellosis/microbiology , Mice, Inbred C57BL , Macrophages/microbiology , Bacterial Proteins/metabolism , Membrane Proteins/metabolism , Endoplasmic Reticulum-Associated Degradation , Ubiquitin-Protein Ligases/metabolism , Endoplasmic Reticulum/microbiology
4.
Proc Natl Acad Sci U S A ; 118(32)2021 08 10.
Article in English | MEDLINE | ID: mdl-34353909

ABSTRACT

Perturbation of the endoplasmic reticulum (ER), a central organelle of the cell, can have critical consequences for cellular homeostasis. An elaborate surveillance system known as ER quality control ensures that cells can respond and adapt to stress via the unfolded protein response (UPR) and that only correctly assembled proteins reach their destination. Interestingly, several bacterial pathogens hijack the ER to establish an infection. However, it remains poorly understood how bacterial pathogens exploit ER quality-control functions to complete their intracellular cycle. Brucella spp. replicate extensively within an ER-derived niche, which evolves into specialized vacuoles suited for exit from infected cells. Here we present Brucella-secreted protein L (BspL), a Brucella abortus effector that interacts with Herp, a central component of the ER-associated degradation (ERAD) machinery. We found that BspL enhances ERAD at the late stages of the infection. BspL targeting of Herp and ERAD allows tight control of the kinetics of autophagic Brucella-containing vacuole formation, delaying the last step of its intracellular cycle and cell-to-cell spread. This study highlights a mechanism by which a bacterial pathogen hijacks ERAD components for fine regulation of its intracellular trafficking.


Subject(s)
Bacterial Proteins/metabolism , Brucella abortus/pathogenicity , Brucellosis/metabolism , Animals , Bacterial Proteins/genetics , Brucella abortus/metabolism , Brucellosis/microbiology , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum-Associated Degradation , HeLa Cells , Host-Pathogen Interactions/physiology , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Inbred C57BL , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Transcription Factor CHOP/genetics , Type IV Secretion Systems/metabolism , X-Box Binding Protein 1/genetics
5.
EMBO J ; 40(19): e107664, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34423453

ABSTRACT

Remodeling of host cellular membrane transport pathways is a common pathogenic trait of many intracellular microbes that is essential to their intravacuolar life cycle and proliferation. The bacterium Brucella abortus generates a host endoplasmic reticulum-derived vacuole (rBCV) that supports its intracellular growth, via VirB Type IV secretion system-mediated delivery of effector proteins, whose functions and mode of action are mostly unknown. Here, we show that the effector BspF specifically promotes Brucella replication within rBCVs by interfering with vesicular transport between the trans-Golgi network (TGN) and recycling endocytic compartment. BspF targeted the recycling endosome, inhibited retrograde traffic to the TGN, and interacted with the Arf6 GTPase-activating Protein (GAP) ACAP1 to dysregulate Arf6-/Rab8a-dependent transport within the recycling endosome, which resulted in accretion of TGN-associated vesicles by rBCVs and enhanced bacterial growth. Altogether, these findings provide mechanistic insight into bacterial modulation of membrane transport used to promote their own proliferation within intracellular vacuoles.


Subject(s)
ADP-Ribosylation Factor 6/metabolism , Brucella abortus/physiology , Brucellosis/metabolism , Brucellosis/microbiology , Host-Pathogen Interactions , Vacuoles/microbiology , rab GTP-Binding Proteins/metabolism , Animals , Bacterial Proteins/metabolism , Brucellosis/immunology , Endosomes/metabolism , Endosomes/microbiology , GTPase-Activating Proteins/metabolism , HeLa Cells , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Mice , Models, Biological , Protein Binding , Protein Transport , Type IV Secretion Systems , trans-Golgi Network
6.
mBio ; 11(2)2020 03 31.
Article in English | MEDLINE | ID: mdl-32234817

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.


Subject(s)
Brucella/physiology , Brucellosis/metabolism , Brucellosis/microbiology , Epistasis, Genetic , Host-Pathogen Interactions , Type IV Secretion Systems/genetics , Type IV Secretion Systems/metabolism , rab2 GTP-Binding Protein/metabolism , Endoplasmic Reticulum/metabolism , Gene Expression Regulation, Bacterial , Golgi Apparatus/metabolism , Macrophages/metabolism , Macrophages/microbiology , Protein Binding , Protein Transport
7.
mBio ; 10(4)2019 07 23.
Article in English | MEDLINE | ID: mdl-31337727

ABSTRACT

Subversion of endoplasmic reticulum (ER) function is a feature shared by multiple intracellular bacteria and viruses, and in many cases this disruption of cellular function activates pathways of the unfolded protein response (UPR). In the case of infection with Brucella abortus, the etiologic agent of brucellosis, the unfolded protein response in the infected placenta contributes to placentitis and abortion, leading to pathogen transmission. Here we show that B. abortus infection of pregnant mice led to death of infected placental trophoblasts in a manner that depended on the VirB type IV secretion system (T4SS) and its effector VceC. The trophoblast death program required the ER stress-induced transcription factor CHOP. While NOD1/NOD2 expression in macrophages contributed to ER stress-induced inflammation, these receptors did not play a role in trophoblast death. Both placentitis and abortion were independent of apoptosis-associated Speck-like protein containing a caspase activation and recruitment domain (ASC). These studies show that B. abortus uses its T4SS to induce cell-type-specific responses to ER stress in trophoblasts that trigger placental inflammation and abortion. Our results suggest further that in B. abortus the T4SS and its effectors are under selection as bacterial transmission factors.IMPORTANCEBrucella abortus infects the placenta of pregnant cows, where it replicates to high levels and triggers abortion of the calf. The aborted material is highly infectious and transmits infection to both cows and humans, but very little is known about how B. abortus causes abortion. By studying this infection in pregnant mice, we discovered that B. abortus kills trophoblasts, which are important cells for maintaining pregnancy. This killing required an injected bacterial protein (VceC) that triggered an endoplasmic reticulum (ER) stress response in the trophoblast. By inhibiting ER stress or infecting mice that lack CHOP, a protein induced by ER stress, we could prevent death of trophoblasts, reduce inflammation, and increase the viability of the pups. Our results suggest that B. abortus injects VceC into placental trophoblasts to promote its transmission by abortion.


Subject(s)
Brucella abortus/pathogenicity , Cell Death , Endoplasmic Reticulum Stress , Placenta/microbiology , Trophoblasts/microbiology , Type IV Secretion Systems/metabolism , Animals , Female , Mice , Mice, Inbred C57BL , Nod1 Signaling Adaptor Protein/genetics , Nod2 Signaling Adaptor Protein/genetics , Placenta/cytology , Pregnancy , Transcription Factor CHOP/genetics , Trophoblasts/pathology , Unfolded Protein Response
8.
PLoS Pathog ; 15(7): e1007959, 2019 07.
Article in English | MEDLINE | ID: mdl-31339948

ABSTRACT

The enteric bacterial pathogen Salmonella enterica serovar Typhimurium (S. Typhimurium), utilizes two type III secretion systems (T3SSs) to invade host cells, survive and replicate intracellularly. T3SS1 and its dedicated effector proteins are required for bacterial entry into non-phagocytic cells and establishment and trafficking of the nascent Salmonella-containing vacuole (SCV). Here we identify the first T3SS1 effector required to maintain the integrity of the nascent SCV as SopF. SopF associates with host cell membranes, either when translocated by bacteria or ectopically expressed. Recombinant SopF binds to multiple phosphoinositides in protein-lipid overlays, suggesting that it targets eukaryotic cell membranes via phospholipid interactions. In yeast, the subcellular localization of SopF is dependent on the activity of Mss4, a phosphatidylinositol 4-phosphate 5-kinase that generates PI(4,5)P2 from PI(4)P, indicating that membrane recruitment of SopF requires specific phospholipids. Ectopically expressed SopF partially colocalizes with specific phosphoinositide pools present on the plasma membrane in mammalian cells and with cytoskeletal-associated markers at the leading edge of cells. Translocated SopF concentrates on plasma membrane ruffles and around intracellular bacteria, presumably on the SCV. SopF is not required for bacterial invasion of non-phagocytic cells but is required for maintenance of the internalization vacuole membrane as infection with a S. Typhimurium ΔsopF mutant led to increased lysis of the SCV compared to wild type bacteria. Our structure-function analysis shows that the carboxy-terminal seven amino acids of SopF are essential for its membrane association in host cells and to promote SCV membrane stability. We also describe that SopF and another T3SS1 effector, SopB, act antagonistically to modulate nascent SCV membrane dynamics. In summary, our study highlights that a delicate balance of type III effector activities regulates the stability of the Salmonella internalization vacuole.


Subject(s)
Salmonella typhimurium/physiology , Type III Secretion Systems/physiology , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Line , Cell Membrane/metabolism , Cell Membrane/microbiology , HeLa Cells , Host Microbial Interactions/physiology , Humans , Mice , Phosphatidylinositols/metabolism , Protein Binding , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Salmonella typhimurium/genetics , Salmonella typhimurium/pathogenicity , Type III Secretion Systems/chemistry , Type III Secretion Systems/genetics , Vacuoles/metabolism , Vacuoles/microbiology
9.
Microbiol Spectr ; 7(2)2019 03.
Article in English | MEDLINE | ID: mdl-30848234

ABSTRACT

Bacteria of the genus Brucella colonize a wide variety of mammalian hosts, in which their infectious cycle and ability to cause disease predominantly rely on an intracellular lifestyle within phagocytes. Upon entry into host cells, Brucella organisms undergo a complex, multistage intracellular cycle in which they sequentially traffic through, and exploit functions of, the endocytic, secretory, and autophagic compartments via type IV secretion system (T4SS)-mediated delivery of bacterial effectors. These effectors modulate an array of host functions and machineries to first promote conversion of the initial endosome-like Brucella-containing vacuole (eBCV) into a replication-permissive organelle derived from the host endoplasmic reticulum (rBCV) and then to an autophagy-related vacuole (aBCV) that mediates bacterial egress. Here we detail and discuss our current knowledge of cellular and molecular events of the Brucella intracellular cycle. We discuss the importance of the endosomal stage in determining T4SS competency, the roles of autophagy in rBCV biogenesis and aBCV formation, and T4SS-driven mechanisms of modulation of host secretory traffic in rBCV biogenesis and bacterial egress.


Subject(s)
Brucella/growth & development , Brucellosis/microbiology , Cytoplasm/microbiology , Animals , Bacterial Proteins/metabolism , Brucella/metabolism , Host-Pathogen Interactions , Humans , Life Cycle Stages , Phagocytes/microbiology , Vacuoles/microbiology
10.
Cell Host Microbe ; 24(2): 285-295.e8, 2018 08 08.
Article in English | MEDLINE | ID: mdl-30057173

ABSTRACT

Many pathogenic intracellular bacteria manipulate the host phago-endosomal system to establish and maintain a permissive niche. The fate and identity of these intracellular compartments is controlled by phosphoinositide lipids. By mechanisms that have remained undefined, a Francisella pathogenicity island-encoded secretion system allows phagosomal escape and replication of bacteria within host cell cytoplasm. Here we report the discovery that a substrate of this system, outside pathogenicity island A (OpiA), represents a family of wortmannin-resistant bacterial phosphatidylinositol (PI) 3-kinase enzymes with members found in a wide range of intracellular pathogens, including Rickettsia and Legionella spp. We show that OpiA acts on the Francisella-containing phagosome and promotes bacterial escape into the cytoplasm. Furthermore, we demonstrate that the phenotypic consequences of OpiA inactivation are mitigated by endosomal maturation arrest. Our findings suggest that Francisella, and likely other intracellular bacteria, override the finely tuned dynamics of phagosomal PI(3)P in order to promote intracellular survival and pathogenesis.


Subject(s)
Francisella/growth & development , Francisella/pathogenicity , Host-Pathogen Interactions/physiology , Phagosomes/metabolism , Phagosomes/microbiology , Phosphatidylinositol 3-Kinase/metabolism , Animals , Bacterial Proteins/metabolism , Cytoplasm/microbiology , DNA Replication , Disease Models, Animal , Endosomes/microbiology , Female , Francisella/genetics , Genes, Bacterial/genetics , Genomic Islands , HEK293 Cells , HeLa Cells , Humans , Lipid Metabolism , Macrophages/microbiology , Male , Mice , Mice, Inbred C57BL , Phosphatidylinositols/metabolism , RAW 264.7 Cells , Type VI Secretion Systems/metabolism , Virulence Factors/metabolism
11.
Cell Host Microbe ; 22(3): 317-329.e7, 2017 Sep 13.
Article in English | MEDLINE | ID: mdl-28844886

ABSTRACT

Many intracellular pathogens exploit host secretory trafficking to support their intracellular cycle, but knowledge of these pathogenic processes is limited. The bacterium Brucella abortus uses a type IV secretion system (VirB T4SS) to generate a replication-permissive Brucella-containing vacuole (rBCV) derived from the host ER, a process that requires host early secretory trafficking. Here we show that the VirB T4SS effector BspB contributes to rBCV biogenesis and Brucella replication by interacting with the conserved oligomeric Golgi (COG) tethering complex, a major coordinator of Golgi vesicular trafficking, thus remodeling Golgi membrane traffic and redirecting Golgi-derived vesicles to the BCV. Altogether, these findings demonstrate that Brucella modulates COG-dependent trafficking via delivery of a T4SS effector to promote rBCV biogenesis and intracellular proliferation, providing mechanistic insight into how bacterial exploitation of host secretory functions promotes pathogenesis.


Subject(s)
Bacterial Proteins/metabolism , Brucella abortus/metabolism , Brucellosis/microbiology , Golgi Apparatus/metabolism , Type IV Secretion Systems/metabolism , Vacuoles/metabolism , Bacterial Proteins/genetics , Brucella abortus/genetics , Brucellosis/metabolism , Cell Line , Golgi Apparatus/microbiology , Host-Pathogen Interactions , Humans , Protein Transport , Type IV Secretion Systems/genetics , Vacuoles/microbiology
12.
mBio ; 7(6)2016 11 29.
Article in English | MEDLINE | ID: mdl-27899503

ABSTRACT

Brucella abortus, the bacterial agent of the worldwide zoonosis brucellosis, primarily infects host phagocytes, where it undergoes an intracellular cycle within a dedicated membrane-bound vacuole, the Brucella-containing vacuole (BCV). Initially of endosomal origin (eBCV), BCVs are remodeled into replication-permissive organelles (rBCV) derived from the host endoplasmic reticulum, a process that requires modulation of host secretory functions via delivery of effector proteins by the Brucella VirB type IV secretion system (T4SS). Following replication, rBCVs are converted into autophagic vacuoles (aBCVs) that facilitate bacterial egress and subsequent infections, arguing that the bacterium sequentially manipulates multiple cellular pathways to complete its cycle. The VirB T4SS is essential for rBCV biogenesis, as VirB-deficient mutants are stalled in eBCVs and cannot mediate rBCV biogenesis. This has precluded analysis of whether the VirB apparatus also drives subsequent stages of the Brucella intracellular cycle. To address this issue, we have generated a B. abortus strain in which VirB T4SS function is conditionally controlled via anhydrotetracycline (ATc)-dependent complementation of a deletion of the virB11 gene encoding the VirB11 ATPase. We show in murine bone marrow-derived macrophages (BMMs) that early VirB production is essential for optimal rBCV biogenesis and bacterial replication. Transient expression of virB11 prior to infection was sufficient to mediate normal rBCV biogenesis and bacterial replication but led to T4SS inactivation and decreased aBCV formation and bacterial release, indicating that these postreplication stages are also T4SS dependent. Hence, our findings support the hypothesis of additional, postreplication roles of type IV secretion in the Brucella intracellular cycle. IMPORTANCE: Many intracellular bacterial pathogens encode specialized secretion systems that deliver effector proteins into host cells to mediate the multiple stages of their intracellular cycles. Because these intracellular events occur sequentially, classical genetic approaches cannot address the late roles that these apparatuses play, as secretion-deficient mutants cannot proceed past their initial defect. Here we have designed a functionally controllable VirB type IV secretion system (T4SS) in the bacterial pathogen Brucella abortus to decipher its temporal requirements during the bacterium's intracellular cycle in macrophages. By controlling production of the VirB11 ATPase, which energizes the T4SS, we show not only that this apparatus is required early to generate the Brucella replicative organelle but also that it contributes to completion of the bacterium's cycle and bacterial egress. Our findings expand upon the pathogenic functions of the Brucella VirB T4SS and illustrate targeting of secretion ATPases as a useful strategy to manipulate the activity of bacterial secretion systems.


Subject(s)
Adenosine Triphosphatases/metabolism , Brucella abortus/metabolism , Host-Pathogen Interactions , Macrophages/microbiology , Type IV Secretion Systems/metabolism , Adenosine Triphosphatases/genetics , Animals , Autophagosomes/metabolism , Autophagosomes/microbiology , Brucella abortus/genetics , Cells, Cultured , Endosomes/metabolism , Endosomes/microbiology , Gene Deletion , Genetic Complementation Test , Mice , Organelle Biogenesis , Type IV Secretion Systems/genetics
13.
Cell Host Microbe ; 20(5): 573-583, 2016 Nov 09.
Article in English | MEDLINE | ID: mdl-27832588

ABSTRACT

The intracellular bacterial pathogen Francisella tularensis causes tularemia, a zoonosis that can be fatal. The type VI secretion system (T6SS) encoded by the Francisella pathogenicity island (FPI) is critical for the virulence of this organism. Existing studies suggest that the complete repertoire of T6SS effectors delivered to host cells is encoded by the FPI. Using a proteome-wide approach, we discovered that the FPI-encoded T6SS exports at least three effectors encoded outside of the island. These proteins share features with virulence determinants of other pathogens, and we provide evidence that they can contribute to intramacrophage growth. The remaining proteins that we identified are encoded within the FPI. Two of these FPI-encoded proteins constitute effectors, whereas the others form a unique complex required for core function of the T6SS apparatus. The discovery of secreted effectors mediating interactions between Francisella and its host significantly advances our understanding of the pathogenesis of this organism.


Subject(s)
Bacterial Proteins/metabolism , Francisella tularensis/growth & development , Francisella tularensis/genetics , Genomic Islands , Host-Pathogen Interactions , Macrophages/microbiology , Virulence Factors/metabolism , Bacterial Proteins/genetics , Protein Transport , Type VI Secretion Systems , Virulence , Virulence Factors/genetics
14.
J Mol Biol ; 428(17): 3387-98, 2016 08 28.
Article in English | MEDLINE | ID: mdl-27456933

ABSTRACT

Autophagy is a conserved lysosomal recycling process, which maintains cellular homeostasis during stress and starvation conditions by degrading and recycling proteins, lipids, and carbohydrates, ultimately increasing nutrient availability in eukaryotes. An additional function of autophagy, termed xenophagy, is to detect, capture, and destroy invading microorganisms, such as viruses, bacteria, and protozoa, providing autophagy with a role in innate immunity. Many intracellular pathogens have, however, developed mechanisms to avoid xenophagy and have evolved strategies to take advantage of select autophagic processes to undergo their intracellular life cycle. This review article will discuss the molecular mechanisms used by the intracellular bacterial pathogens Francisella spp. and Brucella spp. to manipulate components of the autophagic pathway, promoting cytosolic growth in the case of Francisella spp. and facilitating cellular egress and cell-to-cell spread in the case of Brucella spp. These examples highlight how successful, highly infectious bacterial pathogens avoid or subvert host autophagy mechanisms normally employed to maintain eukaryotic homeostasis.


Subject(s)
Autophagy , Brucella/immunology , Brucella/pathogenicity , Francisella/immunology , Francisella/pathogenicity , Host-Pathogen Interactions , Immune Evasion , Animals , Humans
15.
Cell Microbiol ; 17(7): 951-8, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25916795

ABSTRACT

Bacteria of the genus Brucella are intracellular vacuolar pathogens of mammals that cause the worldwide zoonosis brucellosis, and reside within phagocytes of infected hosts to promote their survival, persistence and proliferation. These traits are essential to the bacterium's ability to cause disease and have been the subject of much investigation to gain an understanding of Brucella pathogenic mechanisms. Although the endoplasmic reticulum-derived nature of the Brucella replicative niche has been long known, major strides have recently been made in deciphering the molecular mechanisms of its biogenesis, including the identification of bacterial determinants and host cellular pathways involved in this process. Here I will review and discuss the most recent advances in our knowledge of Brucella intracellular pathogenesis, with an emphasis on bacterial exploitation of the host endoplasmic reticulum-associated functions, and how autophagy-related processes contribute to the bacterium's intracellular cycle.


Subject(s)
Brucella/growth & development , Host-Pathogen Interactions , Phagocytes/microbiology , Vacuoles/metabolism , Vacuoles/microbiology , Virulence Factors/metabolism , Animals , Brucella/immunology , Humans , Mammals , Models, Biological , Phagocytes/immunology
16.
Nat Rev Microbiol ; 13(2): 71-82, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25534809

ABSTRACT

The unfolded protein response (UPR) is a cytoprotective response that is aimed at restoring cellular homeostasis following physiological stress exerted on the endoplasmic reticulum (ER), which also invokes innate immune signalling in response to invading microorganisms. Although it has been known for some time that the UPR is modulated by various viruses, recent evidence indicates that it also has multiple roles during bacterial infections. In this Review, we describe how bacteria interact with the ER, including how bacteria induce the UPR, how subversion of the UPR promotes bacterial proliferation and how the UPR contributes to innate immune responses against invading bacteria.


Subject(s)
Bacteria , Endoplasmic Reticulum/microbiology , Endoplasmic Reticulum/physiology , Immunity, Innate/physiology , Unfolded Protein Response/physiology , Animals , Bacteria/immunology , Bacterial Infections/immunology , Cell Proliferation
17.
Cell Host Microbe ; 16(2): 249-256, 2014 Aug 13.
Article in English | MEDLINE | ID: mdl-25121752

ABSTRACT

Inflammasome-mediated host defenses have been extensively studied in innate immune cells. Whether inflammasomes function for innate defense in intestinal epithelial cells, which represent the first line of defense against enteric pathogens, remains unknown. We observed enhanced Salmonella enterica serovar Typhimurium colonization in the intestinal epithelium of caspase-11-deficient mice, but not at systemic sites. In polarized epithelial monolayers, siRNA-mediated depletion of caspase-4, a human ortholog of caspase-11, also led to increased bacterial colonization. Decreased rates of pyroptotic cell death, a host defense mechanism that extrudes S. Typhimurium-infected cells from the polarized epithelium, accounted for increased pathogen burdens. The caspase-4 inflammasome also governs activation of the proinflammatory cytokine, interleukin (IL)-18, in response to intracellular (S. Typhimurium) and extracellular (enteropathogenic Escherichia coli) enteric pathogens, via intracellular LPS sensing. Therefore, an epithelial cell-intrinsic noncanonical inflammasome plays a critical role in antimicrobial defense at the intestinal mucosal surface.


Subject(s)
Caspases, Initiator/metabolism , Caspases/metabolism , Escherichia coli Infections/enzymology , Inflammasomes/physiology , Salmonella Infections/enzymology , Animals , Cell Line, Tumor , Enteropathogenic Escherichia coli/immunology , Enzyme Activation , Escherichia coli Infections/immunology , Gastroenteritis/enzymology , Gastroenteritis/microbiology , Humans , Interleukin-18/metabolism , Intestinal Mucosa/enzymology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Lipopolysaccharides/pharmacology , Mice, Inbred C57BL , Mice, Knockout , Salmonella Infections/immunology , Salmonella enterica/immunology
18.
Infect Immun ; 82(7): 2935-48, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24778115

ABSTRACT

The Francisella FTT0831c/FTL_0325 gene encodes amino acid motifs to suggest it is a lipoprotein and that it may interact with the bacterial cell wall as a member of the OmpA-like protein family. Previous studies have suggested that FTT0831c is surface exposed and required for virulence of Francisella tularensis by subverting the host innate immune response (M. Mahawar et al., J. Biol. Chem. 287:25216-25229, 2012). We also found that FTT0831c is required for murine pathogenesis and intramacrophage growth of Schu S4, but we propose a different model to account for the proinflammatory nature of the resultant mutants. First, inactivation of FTL_0325 from live vaccine strain (LVS) or FTT0831c from Schu S4 resulted in temperature-dependent defects in cell viability and morphology. Loss of FTT0831c was also associated with an unusual defect in lipopolysaccharide O-antigen synthesis, but loss of FTL_0325 was not. Full restoration of these properties was observed in complemented strains expressing FTT0831c in trans, but not in strains lacking the OmpA motif, suggesting that cell wall contact is required. Finally, growth of the LVS FTL_0325 mutant in Mueller-Hinton broth at 37°C resulted in the appearance of membrane blebs at the poles and midpoint, prior to the formation of enlarged round cells that showed evidence of compromised cellular membranes. Taken together, these data are more consistent with the known structural role of OmpA-like proteins in linking the OM to the cell wall and, as such, maintenance of structural integrity preventing altered surface exposure or release of Toll-like receptor 2 agonists during rapid growth of Francisella in vitro and in vivo.


Subject(s)
Bacterial Proteins/metabolism , Francisella tularensis/cytology , Francisella tularensis/physiology , Gene Expression Regulation, Bacterial/physiology , Animals , Bacterial Proteins/genetics , Cell Shape , Female , Francisella tularensis/genetics , Gene Deletion , Genetic Complementation Test , Immunity, Innate , Mice , Mice, Inbred C3H , Tularemia/microbiology
19.
Cell Microbiol ; 16(6): 862-77, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24286610

ABSTRACT

Autophagy is a key innate immune response to intracellular parasites that promotes their delivery to degradative lysosomes following detection in the cytosol or within damaged vacuoles. Like Listeria and Shigella, which use specific mechanisms to avoid autophagic detection and capture, the bacterial pathogen Francisella tularensis proliferates within the cytosol of macrophages without demonstrable control by autophagy. To examine how Francisella evades autophagy, we screened a library of F. tularensis subsp. tularensis Schu S4 HimarFT transposon mutants in GFP-LC3-expressing murine macrophages by microscopy for clones localized within autophagic vacuoles after phagosomal escape. Eleven clones showed autophagic capture at 6 h post-infection, whose HimarFT insertions clustered to fourgenetic loci involved in lipopolysaccharidic and capsular O-antigen biosynthesis. Consistent with the HimarFT mutants, in-frame deletion mutants of two representative loci, FTT1236 and FTT1448c (manC), lacking both LPS and capsular O-antigen, underwent phagosomal escape but were cleared from the host cytosol. Unlike wild-type Francisella, the O-antigen deletion mutants were ubiquitinated, and recruited the autophagy adaptor p62/SQSTM1 and LC3 prior to cytosolic clearance. Autophagy-deficient macrophages partially supported replication of both mutants, indicating that O-antigen-lacking Francisella are controlled by autophagy. These data demonstrate the intracellular protective role of this bacterial surface polysaccharide against autophagy.


Subject(s)
Autophagy , Francisella tularensis/immunology , Macrophages/immunology , Macrophages/microbiology , Microbial Viability , O Antigens/immunology , O Antigens/metabolism , Animals , Cells, Cultured , Cytosol/microbiology , DNA Transposable Elements , Francisella tularensis/physiology , Host-Pathogen Interactions , Mice, Inbred C57BL , Mutagenesis, Insertional
20.
PLoS Pathog ; 9(8): e1003556, 2013.
Article in English | MEDLINE | ID: mdl-23950720

ABSTRACT

The intracellular pathogenic bacterium Brucella generates a replicative vacuole (rBCV) derived from the endoplasmic reticulum via subversion of the host cell secretory pathway. rBCV biogenesis requires the expression of the Type IV secretion system (T4SS) VirB, which is thought to translocate effector proteins that modulate membrane trafficking along the endocytic and secretory pathways. To date, only a few T4SS substrates have been identified, whose molecular functions remain unknown. Here, we used an in silico screen to identify putative T4SS effector candidate proteins using criteria such as limited homology in other bacterial genera, the presence of features similar to known VirB T4SS effectors, GC content and presence of eukaryotic-like motifs. Using ß-lactamase and CyaA adenylate cyclase reporter assays, we identified eleven proteins translocated into host cells by Brucella, five in a VirB T4SS-dependent manner, namely BAB1_0678 (BspA), BAB1_0712 (BspB), BAB1_0847 (BspC), BAB1_1671 (BspE) and BAB1_1948 (BspF). A subset of the translocated proteins targeted secretory pathway compartments when ectopically expressed in HeLa cells, and the VirB effectors BspA, BspB and BspF inhibited protein secretion. Brucella infection also impaired host protein secretion in a process requiring BspA, BspB and BspF. Single or combined deletions of bspA, bspB and bspF affected Brucella ability to replicate in macrophages and persist in the liver of infected mice. Taken together, these findings demonstrate that Brucella modulates secretory trafficking via multiple T4SS effector proteins that likely act coordinately to promote Brucella pathogenesis.


Subject(s)
Bacterial Proteins/metabolism , Bacterial Secretion Systems/physiology , Brucella abortus/metabolism , Brucellosis/metabolism , Liver/metabolism , Macrophages/metabolism , Macrophages/microbiology , Membrane Proteins/metabolism , Animals , Bacterial Proteins/genetics , Brucella abortus/genetics , Brucellosis/pathology , Female , HeLa Cells , Humans , Liver/microbiology , Liver/pathology , Macrophages/pathology , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Protein Transport/physiology , Vacuoles/genetics , Vacuoles/metabolism , Vacuoles/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL
...