Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Front Immunol ; 14: 1021824, 2023.
Article in English | MEDLINE | ID: mdl-37153622

ABSTRACT

Mucosal delivery of IL-27 has been shown to have a therapeutic benefit in murine models of inflammatory bowel disease (IBD). The IL-27 effect was associated with phosphorylated STAT1 (pSTAT1), a product of IL27 receptor signaling, in bowel tissue. To determine whether IL-27 acted directly on colonic epithelium, murine colonoids and primary intact colonic crypts were shown to be unresponsive to IL-27 in vitro and to lack detectable IL-27 receptors. On the other hand, macrophages, which are present in inflamed colon tissue, were responsive to IL-27 in vitro. IL-27 induced pSTAT1 in macrophages, the transcriptome indicated an IFN-like signature, and supernatants induced pSTAT1 in colonoids. IL-27 induced anti-viral activity in macrophages and MHC Class II induction. We conclude that the effects of mucosal delivery of IL-27 in murine IBD are in part based on the known effects of IL27 inducing immunosuppression of T cells mediated by IL-10. We also conclude that IL-27 has potent effects on macrophages in inflamed colon tissue, generating mediators that in turn act on colonic epithelium.


Subject(s)
Inflammatory Bowel Diseases , Interleukin-27 , Mice , Animals , Interleukin-27/therapeutic use , Colon , Inflammatory Bowel Diseases/drug therapy , Macrophages , Epithelium
3.
PLoS Pathog ; 17(3): e1009395, 2021 03.
Article in English | MEDLINE | ID: mdl-33684179

ABSTRACT

The mammalian immune system is constantly challenged by signals from both pathogenic and non-pathogenic microbes. Many of these non-pathogenic microbes have pathogenic potential if the immune system is compromised. The importance of type I interferons (IFNs) in orchestrating innate immune responses to pathogenic microbes has become clear in recent years. However, the control of opportunistic pathogens-and especially intracellular bacteria-by type I IFNs remains less appreciated. In this study, we use the opportunistic, Gram-negative bacterial pathogen Burkholderia cenocepacia (Bc) to show that type I IFNs are capable of limiting bacterial replication in macrophages, preventing illness in immunocompetent mice. Sustained type I IFN signaling through cytosolic receptors allows for increased expression of autophagy and linear ubiquitination mediators, which slows bacterial replication. Transcriptomic analyses and in vivo studies also show that LPS stimulation does not replicate the conditions of intracellular Gram-negative bacterial infection as it pertains to type I IFN stimulation or signaling. This study highlights the importance of type I IFNs in protection against opportunistic pathogens through innate immunity, without the need for damaging inflammatory responses.


Subject(s)
Burkholderia Infections/immunology , Burkholderia cenocepacia/immunology , Immunity, Innate/immunology , Interferon Type I/immunology , Macrophages/immunology , Animals , Cytosol/immunology , Cytosol/microbiology , Female , Male , Mice , Mice, Inbred C57BL
4.
Nat Commun ; 12(1): 348, 2021 01 13.
Article in English | MEDLINE | ID: mdl-33441540

ABSTRACT

In the enteric pathogen Salmonella enterica serovar Typhimurium, invasion and motility are coordinated by the master regulator HilD, which induces expression of the type III secretion system 1 (T3SS1) and motility genes. Methyl-accepting chemotaxis proteins (MCPs) detect specific ligands and control the direction of the flagellar motor, promoting tumbling and changes in direction (if a repellent is detected) or smooth swimming (in the presence of an attractant). Here, we show that HilD induces smooth swimming by upregulating an uncharacterized MCP (McpC), and this is important for invasion of epithelial cells. Remarkably, in vitro assays show that McpC can suppress tumbling and increase smooth swimming in the absence of exogenous ligands. Expression of mcpC is repressed by the universal regulator H-NS, which can be displaced by HilD. Our results highlight the importance of smooth swimming for Salmonella Typhimurium invasiveness and indicate that McpC can act via a ligand-independent mechanism when incorporated into the chemotactic receptor array.


Subject(s)
Bacterial Proteins/metabolism , Chemotaxis/physiology , Methyl-Accepting Chemotaxis Proteins/metabolism , Salmonella typhimurium/metabolism , Transcription Factors/metabolism , Animals , Bacterial Proteins/genetics , Caco-2 Cells , Cattle , Cells, Cultured , Chemotaxis/genetics , Gene Expression Regulation, Bacterial , HeLa Cells , Humans , Methyl-Accepting Chemotaxis Proteins/genetics , Mice, Inbred C57BL , Movement/physiology , Mutation , Salmonella Infections/microbiology , Salmonella typhimurium/genetics , Salmonella typhimurium/physiology , Transcription Factors/genetics
5.
Article in English | MEDLINE | ID: mdl-31482073

ABSTRACT

Numerous methods exist for fluorescently labeling proteins either as direct fusion proteins (GFP, RFP, YFP, etc.-attached to the protein of interest) or utilizing accessory proteins to produce fluorescence (SNAP-tag, CLIP-tag), but the significant increase in size that these accompanying proteins add may hinder or impede proper protein folding, cellular localization, or oligomerization. Fluorescently labeling proteins with biarsenical dyes, like FlAsH, circumvents this issue by using a short 6-amino acid tetracysteine motif that binds the membrane-permeable dye and allows visualization of living cells. Here, we report the successful adaptation of FlAsH dye for live-cell imaging of two genera of spirochetes, Leptospira and Borrelia, by labeling inner or outer membrane proteins tagged with tetracysteine motifs. Visualization of labeled spirochetes was possible by fluorescence microscopy and flow cytometry. A subsequent increase in fluorescent signal intensity, including prolonged detection, was achieved by concatenating two copies of the 6-amino acid motif. Overall, we demonstrate several positive attributes of the biarsenical dye system in that the technique is broadly applicable across spirochete genera, the tetracysteine motif is stably retained and does not interfere with protein function throughout the B. burgdorferi infectious cycle, and the membrane-permeable nature of the dyes permits fluorescent detection of proteins in different cellular locations without the need for fixation or permeabilization. Using this method, new avenues of investigation into spirochete morphology and motility, previously inaccessible with large fluorescent proteins, can now be explored.


Subject(s)
Bacterial Proteins/metabolism , Fluorescent Dyes , Membrane Proteins/metabolism , Microscopy, Fluorescence , Spirochaetales/cytology , Spirochaetales/metabolism , Staining and Labeling , Animals , Bacterial Proteins/genetics , Flow Cytometry , Genes, Bacterial , Humans , Membrane Proteins/genetics , Mice , Spirochaetales/genetics , Spirochaetales Infections/microbiology
6.
Mol Microbiol ; 112(4): 1270-1283, 2019 10.
Article in English | MEDLINE | ID: mdl-31370104

ABSTRACT

Salmonella enterica serovar Typhimurium is a facultative intracellular pathogen that invades the intestinal epithelium. Following invasion of epithelial cells, Salmonella survives and replicates within two distinct intracellular niches. While all of the bacteria are initially taken up into a membrane bound vacuole, the Salmonella-containing vacuole or SCV, a significant proportion of them promptly escape into the cytosol. Cytosolic Salmonella replicates more rapidly compared to the vacuolar population, although the reasons for this are not well understood. SipA, a multi-function effector protein, has been shown to affect intracellular replication and is secreted by cytosolic Salmonella via the invasion-associated Type III Secretion System 1 (T3SS1). Here, we have used a multipronged microscopy approach to show that SipA does not affect bacterial replication rates per se, but rather mediates intra-cytosolic survival and/or initiation of replication following bacterial egress from the SCV. Altogether, our findings reveal an important role for SipA in the early survival of cytosolic Salmonella.


Subject(s)
Bacterial Proteins/metabolism , Epithelial Cells/metabolism , Microfilament Proteins/metabolism , Type III Secretion Systems/metabolism , Adaptation, Physiological/physiology , Bacteria/metabolism , Bacterial Proteins/physiology , Cytoplasm/metabolism , Cytosol/metabolism , Cytosol/physiology , Epithelial Cells/physiology , HeLa Cells , Humans , Microfilament Proteins/physiology , Salmonella Infections/microbiology , Salmonella enterica/metabolism , Salmonella typhimurium/metabolism , Type III Secretion Systems/physiology , Vacuoles/physiology
7.
Curr Protoc Microbiol ; 50(1): e56, 2018 08.
Article in English | MEDLINE | ID: mdl-29927091

ABSTRACT

The successful infection of macrophages by non-typhoidal serovars of Salmonella enterica is likely essential to the establishment of the systemic disease they sometimes cause in susceptible human populations. However, the interactions between Salmonella and human macrophages are not widely studied, with mouse macrophages being a much more common model system. Fundamental differences between mouse and human macrophages make this less than ideal. Additionally, the inability of human macrophage-like cell lines to replicate some properties of primary macrophages makes the use of primary cells desirable. Here we present protocols to study the infection of human monocyte-derived macrophages with Salmonella Typhimurium. These include a method for differentiating monocyte-derived macrophages in vitro and protocols for infecting them with Salmonella Typhimurium, as well as assays to measure the extent of infection, replication, and death. These protocols are useful for the investigation of both bacterial and host factors that determine the outcome of infection. © 2018 by John Wiley & Sons, Inc.


Subject(s)
Cell Culture Techniques/methods , Cell Separation/methods , Macrophages/microbiology , Microscopy/methods , Monocytes/microbiology , Salmonella Infections/microbiology , Salmonella typhimurium/physiology , Animals , Cell Differentiation , Cells, Cultured , Humans , Intestines/microbiology , Macrophages/cytology , Mice , Monocytes/cytology , Salmonella typhimurium/genetics , Salmonella typhimurium/growth & development
8.
PLoS One ; 13(3): e0193601, 2018.
Article in English | MEDLINE | ID: mdl-29538403

ABSTRACT

THP-1 cells differentiated with phorbol 12-myristate 13-acetate (PMA) are widely used as a model for function and biology of human macrophages. However, the conditions used for differentiation, particularly the concentration of PMA and the duration of treatment, vary widely. Here we compare several differentiation conditions and compare the ability of THP-1 macrophages to interact with the facultative intracellular pathogen Salmonella enterica serovar Typhimurium. The results show that THP-1 macrophages differentiated in high concentrations of PMA rapidly died following infection whereas those differentiated in low concentrations of PMA survived and were able to control the intracellular bacteria similar to primary human macrophages.


Subject(s)
Cell Differentiation/drug effects , Salmonella typhimurium/physiology , Tetradecanoylphorbol Acetate/pharmacology , Apoptosis/drug effects , CD11 Antigens/metabolism , Cell Line , Humans , Lipopolysaccharide Receptors/metabolism , Lipopolysaccharides/toxicity , Macrophages/cytology , Macrophages/metabolism , Macrophages/microbiology
9.
mSphere ; 3(1)2018.
Article in English | MEDLINE | ID: mdl-29299535

ABSTRACT

Neutrophils are essential cells of host innate immunity. Although the role of neutrophils in defense against bacterial and fungal infections is well characterized, there is a relative paucity of information about their role against viral infections. Influenza A virus (IAV) infection can be associated with secondary bacterial coinfection, and it has long been posited that the ability of IAV to alter normal neutrophil function predisposes individuals to secondary bacterial infections. To better understand this phenomenon, we evaluated the interaction of pandemic or seasonal H1N1 IAV with human neutrophils isolated from healthy persons. These viruses were ingested by human neutrophils and elicited changes in neutrophil gene expression that are consistent with an interferon-mediated immune response. The viability of neutrophils following coculture with either pandemic or seasonal H1N1 IAV was similar for up to 18 h of culture. Notably, neutrophil exposure to seasonal (but not pandemic) IAV primed these leukocytes for enhanced functions, including production of reactive oxygen species and bactericidal activity. Taken together, our results are at variance with the universal idea that IAV impairs neutrophil function directly to predispose individuals to secondary bacterial infections. Rather, we suggest that some strains of IAV prime neutrophils for enhanced bacterial clearance. IMPORTANCE A long-standing notion is that IAV inhibits normal neutrophil function and thereby predisposes individuals to secondary bacterial infections. Here we report that seasonal H1N1 IAV primes human neutrophils for enhanced killing of Staphylococcus aureus. Moreover, we provide a comprehensive view of the changes in neutrophil gene expression during interaction with seasonal or pandemic IAV and report how these changes relate to functions such as bactericidal activity. This study expands our knowledge of IAV interactions with human neutrophils.

10.
Article in English | MEDLINE | ID: mdl-29201859

ABSTRACT

Here we describe the use of synthetic genetic elements to improve the predictability and tunability of episomal protein production in Salmonella. We used a multi-pronged approach, in which a series of variable-strength synthetic promoters were combined with a synthetic transcriptional terminator, and plasmid copy number variation. This yielded a series of plasmids that drive uniform production of fluorescent and endogenous proteins, over a wide dynamic range. We describe several examples where this system is used to fine-tune constitutive expression in Salmonella, providing an efficient means to titrate out toxic effects of protein production.


Subject(s)
Genes, Bacterial/genetics , Host-Pathogen Interactions/genetics , Plasmids/genetics , Promoter Regions, Genetic/genetics , Salmonella/genetics , Salmonella/metabolism , Bacterial Proteins/genetics , Cytosol , DNA Copy Number Variations , Green Fluorescent Proteins , HeLa Cells , Humans , Salmonella/pathogenicity , Salmonella Infections/genetics , Salmonella Infections/metabolism , Salmonella enterica , Trans-Activators/genetics , Type III Secretion Systems/genetics
11.
PLoS Pathog ; 13(4): e1006354, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28426838

ABSTRACT

Type III secretion system 1 (T3SS1) is used by the enteropathogen Salmonella enterica serovar Typhimurium to establish infection in the gut. Effector proteins translocated by this system across the plasma membrane facilitate invasion of intestinal epithelial cells. One such effector, the inositol phosphatase SopB, contributes to invasion and mediates activation of the pro-survival kinase Akt. Following internalization, some bacteria escape from the Salmonella-containing vacuole into the cytosol and there is evidence suggesting that T3SS1 is expressed in this subpopulation. Here, we investigated the post-invasion role of T3SS1, using SopB as a model effector. In cultured epithelial cells, SopB-dependent Akt phosphorylation was observed at two distinct stages of infection: during and immediately after invasion, and later during peak cytosolic replication. Single cell analysis revealed that cytosolic Salmonella deliver SopB via T3SS1. Although intracellular replication was unaffected in a SopB deletion mutant, cells infected with ΔsopB demonstrated a lack of Akt phosphorylation, earlier time to death, and increased lysis. When SopB expression was induced specifically in cytosolic Salmonella, these effects were restored to levels observed in WT infected cells, indicating that the second wave of SopB protects this infected population against cell death via Akt activation. Thus, T3SS1 has two, temporally distinct roles during epithelial cell colonization. Additionally, we found that delivery of SopB by cytosolic bacteria was translocon-independent, in contrast to canonical effector translocation across eukaryotic membranes, which requires formation of a translocon pore. This mechanism was also observed for another T3SS1 effector, SipA. These findings reveal the functional and mechanistic adaptability of a T3SS that can be harnessed in different microenvironments.


Subject(s)
Bacterial Proteins/metabolism , Epithelial Cells/microbiology , Proto-Oncogene Proteins c-akt/metabolism , Salmonella Infections/microbiology , Salmonella Infections/pathology , Salmonella typhimurium/physiology , Type III Secretion Systems/metabolism , Animals , Bacterial Proteins/genetics , DNA Replication , Epithelial Cells/physiology , Humans , Mice , Phosphorylation , Proto-Oncogene Proteins c-akt/genetics , Salmonella typhimurium/pathogenicity , Type III Secretion Systems/genetics
12.
PLoS One ; 12(3): e0173528, 2017.
Article in English | MEDLINE | ID: mdl-28278296

ABSTRACT

Published data show that murine bone marrow-derived macrophages (BMDM) restrict growth of avirulent phase II, but not virulent phase I, Coxiella burnetii. Growth restriction of phase II bacteria is thought to result from potentiated recognition of pathogen-associated molecular patterns, which leads to production of inhibitory effector molecules. Past studies have used conditioned medium from L-929 murine fibroblasts as a source of macrophage-colony stimulating factor (M-CSF) to promote differentiation of bone marrow-derived myeloid precursors into macrophages. However, uncharacterized components of conditioned medium, such as variable amounts of type I interferons, can affect macrophage activation status and their permissiveness for infection. In the current study, we show that the C. burnetii Nine Mile phase II (NMII) strain grows robustly in primary macrophages from C57BL/6J mice when bone marrow cells are differentiated with recombinant murine M-CSF (rmM-CSF). Bacteria were readily internalized by BMDM, and replicated within degradative, LAMP1-positive vacuoles to achieve roughly 3 logs of growth over 6 days. Uninfected BMDM did not appreciably express CD38 or Egr2, markers of classically (M1) and alternatively (M2) activated macrophages, respectively, nor did infection change the lack of polarization. In accordance with an M0 phenotype, infected BMDM produced moderate amounts of TNF and nitric oxide. Similar NMII growth results were obtained using C57BL/6J myeloid progenitors immortalized with an estrogen-regulated Hoxb8 (ER-Hoxb8) oncogene. To demonstrate the utility of the ER-Hoxb8 system, myeloid progenitors from natural resistance-associated macrophage protein 1 (Nramp1) C57BL/6J knock-in mice were transduced with ER-Hoxb8, and macrophages were derived from immortalized progenitors using rmM-CSF and infected with NMII. No difference in growth was observed when compared to macrophages from wild type mice, indicating depletion of metal ions by the Nramp1 transporter does not negatively impact NMII growth. Results with NMII were recapitulated in primary macrophages where C57BL/6J Nramp1+ BMDM efficiently killed Salmonella enterica serovar Typhimurium. M-CSF differentiated murine macrophages from bone marrow and conditional ER-Hoxb8 myeloid progenitors will be useful ex vivo models for studying Coxiella-macrophage interactions.


Subject(s)
Bone Marrow/microbiology , Coxiella burnetii/growth & development , Macrophages/microbiology , Q Fever/microbiology , Animals , Bone Marrow/metabolism , Cells, Cultured , Colony-Stimulating Factors/metabolism , Coxiella burnetii/pathogenicity , Female , Macrophages/cytology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Q Fever/metabolism , Q Fever/pathology , Tumor Necrosis Factor-alpha/metabolism
13.
Am J Pathol ; 187(1): 187-199, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27955815

ABSTRACT

In the current study, we examined the ability of Salmonella enterica serovar Typhimurium to infect the central nervous system and cause meningitis following the natural route of infection in mice. C57BL/6J mice are extremely susceptible to systemic infection by Salmonella Typhimurium because of loss-of-function mutations in Nramp1 (SLC11A1), a phagosomal membrane protein that controls iron export from vacuoles and inhibits Salmonella growth in macrophages. Therefore, we assessed the ability of Salmonella to disseminate to the central nervous system (CNS) after oral infection in C57BL/6J mice expressing either wild-type (resistant) or mutant (susceptible) alleles of Nramp1. In both strains, oral infection resulted in focal meningitis and ventriculitis with recruitment of inflammatory monocytes to the CNS. In susceptible Nramp1-/- mice, there was a direct correlation between bacteremia and the number of bacteria in the brain, which was not observed in resistant Nramp1+/+ mice. A small percentage of Nramp1+/+ mice developed severe ataxia, which was associated with high bacterial loads in the CNS as well as clear histopathology of necrotizing vasculitis and hemorrhage in the brain. Thus, Nramp1 is not essential for Salmonella entry into the CNS or neuroinflammation, but may influence the mechanisms of CNS entry as well as the severity of meningitis.


Subject(s)
Cell Movement , Meningitis/microbiology , Meningitis/pathology , Monocytes/pathology , Salmonella typhimurium/physiology , Administration, Oral , Animals , Ataxia/metabolism , Ataxia/pathology , Bacteremia/complications , Bacteremia/microbiology , Bacteremia/pathology , Brain/microbiology , Brain/pathology , Cation Transport Proteins/deficiency , Cation Transport Proteins/metabolism , Cerebral Ventricles/pathology , Colony Count, Microbial , Encephalitis/complications , Encephalitis/metabolism , Encephalitis/pathology , Immunohistochemistry , Macrophages/pathology , Meningitis/complications , Mice, Inbred C57BL , Neutrophil Infiltration , Salmonella Infections, Animal/complications , Salmonella Infections, Animal/microbiology , Salmonella Infections, Animal/pathology
14.
mBio ; 6(6): e01648-15, 2015 Nov 10.
Article in English | MEDLINE | ID: mdl-26556273

ABSTRACT

UNLABELLED: Chlamydia trachomatis is an obligate intracellular bacterium that is a globally important human pathogen. The chlamydial plasmid is an attenuating virulence factor, but the molecular basis for attenuation is not understood. Chlamydiae replicate within a membrane-bound vacuole termed an inclusion, where they undergo a biphasic developmental growth cycle and differentiate from noninfectious into infectious organisms. Late in the developmental cycle, the fragile chlamydia-laden inclusion retains its integrity by surrounding itself with scaffolds of host cytoskeletal proteins. The ability of chlamydiae to developmentally free themselves from this cytoskeleton network is a fundamental virulence trait of the pathogen. Here, we show that plasmidless chlamydiae are incapable of disrupting their cytoskeletal entrapment and remain intracellular as stable mature inclusions that support high numbers of infectious organisms. By using deletion mutants of the eight plasmid-carried genes (Δpgp1 to Δpgp8), we show that Pgp4, a transcriptional regulator of multiple chromosomal genes, is required for exit. Exit of chlamydiae is dependent on protein synthesis and is inhibited by the compound C1, an inhibitor of the type III secretion system (T3S). Exit of plasmid-free and Δpgp4 organisms, which failed to lyse infected cells, was rescued by latrunculin B, an inhibitor of actin polymerization. Our findings describe a genetic mechanism of chlamydial exit from host cells that is dependent on an unknown pgp4-regulated chromosomal T3S effector gene. IMPORTANCE: Chlamydia's obligate intracellular life style requires both entry into and exit from host cells. Virulence factors that function in exiting are unknown. The chlamydial inclusion is stabilized late in the infection cycle by F-actin. A prerequisite of chlamydial exit is its ability to disassemble actin from the inclusion. We show that chlamydial plasmid-free organisms, and also a plasmid gene protein 4 (pgp4) null mutant, do not disassociate actin from the inclusion and fail to exit cells. We further provide evidence that Pgp4-regulated exit is dependent on the chlamydial type III secretion system. This study is the first to define a genetic mechanism that functions in chlamydial lytic exit from host cells. The findings also have practical implications for understanding why plasmid-free chlamydiae are highly attenuated and have the ability to elicit robust protective immune responses.


Subject(s)
Chlamydia trachomatis/physiology , Epithelial Cells/microbiology , Host-Pathogen Interactions , Plasmids , Vacuoles/microbiology , Actins/metabolism , Chlamydia trachomatis/genetics , Chlamydia trachomatis/growth & development , HeLa Cells , Humans , Virulence , Virulence Factors/metabolism
15.
Infect Immun ; 83(7): 2661-71, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25895967

ABSTRACT

Salmonella enterica serovar Typhimurium is a common cause of food-borne gastrointestinal illness, but additionally it causes potentially fatal bacteremia in some immunocompromised patients. In mice, systemic spread and replication of the bacteria depend upon infection of and replication within macrophages, but replication in human macrophages is not widely reported or well studied. In order to assess the ability of Salmonella Typhimurium to replicate in human macrophages, we infected primary monocyte-derived macrophages (MDM) that had been differentiated under conditions known to generate different phenotypes. We found that replication in MDM depends greatly upon the phenotype of the cells, as M1-skewed macrophages did not allow replication, while M2a macrophages and macrophages differentiated with macrophage colony-stimulating factor (M-CSF) alone (termed M0) did. We describe how additional conditions that alter the macrophage phenotype or the gene expression of the bacteria affect the outcome of infection. In M0 MDM, the temporal expression of representative genes from Salmonella pathogenicity islands 1 and 2 (SPI1 and SPI2) and the importance of the PhoP/Q two-component regulatory system are similar to what has been shown in mouse macrophages. However, in contrast to mouse macrophages, where replication is SPI2 dependent, we observed early SPI2-independent replication in addition to later SPI2-dependent replication in M0 macrophages. Only SPI2-dependent replication was associated with death of the host cell at later time points. Altogether, our results reveal a very nuanced interaction between Salmonella and human macrophages.


Subject(s)
Host-Pathogen Interactions , Macrophages/immunology , Macrophages/microbiology , Salmonella typhimurium/growth & development , Cell Survival , Cells, Cultured , Genomic Islands , Humans , Salmonella typhimurium/genetics
16.
J Leukoc Biol ; 95(3): 389-98, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24227798

ABSTRACT

Yersinia pestis, the bacterial agent of plague, is transmitted by fleas. The bite of an infected flea deposits Y. pestis into the dermis and triggers recruitment of innate immune cells, including phagocytic PMNs. Y. pestis can subvert this PMN response and survive at the flea-bite site, disseminate, and persist in the host. Although its genome encodes a number of antiphagocytic virulence factors, phagocytosis of Y. pestis by PMNs has been observed. This study tests the hypotheses that Y. pestis, grown at the ambient temperature of the flea vector (21°C), where the major antiphagocytic virulence factors are not produced, can survive and replicate within human PMNs and can use PMNs as a route to infect macrophages subsequently. We show that Y. pestis is localized within PMN phagosomes, predominately as individual bacteria, and that intracellular bacteria can survive and replicate. Within 12 h of infection, ~70% of infected PMNs had PS on their surface and were plausibly competent for efferocytosis. With the use of live cell confocal imaging, we show that autologous HMDMs recognize and internalize infected PMNs and that Y. pestis survives and replicates within these HMDMs following efferocytosis. Addition of HMDMs to infected PMNs resulted in decreased secretion of inflammatory cytokines (compared with HMDMs incubated directly with pCD1(-) Y. pestis) and increased secretion of the anti-inflammatory cytokine IL-1ra. Thus, Y. pestis can survive and replicate within PMNs, and infected PMNs may be a route for noninflammatory infection of macrophages.


Subject(s)
Macrophages/microbiology , Neutrophils/microbiology , Phagosomes/microbiology , Plague/microbiology , Yersinia pestis/pathogenicity , Humans , Macrophages/immunology , Microscopy, Confocal , Microscopy, Electron, Transmission , Neutrophils/immunology , Phagosomes/immunology , Virulence , Yersinia pestis/immunology
17.
mBio ; 4(1): e00418-12, 2013 Feb 19.
Article in English | MEDLINE | ID: mdl-23422410

ABSTRACT

Host cytokine responses to Brucella abortus infection are elicited predominantly by the deployment of a type IV secretion system (T4SS). However, the mechanism by which the T4SS elicits inflammation remains unknown. Here we show that translocation of the T4SS substrate VceC into host cells induces proinflammatory responses. Ectopically expressed VceC interacted with the endoplasmic reticulum (ER) chaperone BiP/Grp78 and localized to the ER of HeLa cells. ER localization of VceC required a transmembrane domain in its N terminus. Notably, the expression of VceC resulted in reorganization of ER structures. In macrophages, VceC was required for B. abortus-induced inflammation by induction of the unfolded protein response by a process requiring inositol-requiring transmembrane kinase/endonuclease 1. Altogether, these findings suggest that translocation of the T4SS effector VceC induces ER stress, which results in the induction of proinflammatory host cell responses during B. abortus infection. IMPORTANCE Brucella species are pathogens that require a type IV secretion system (T4SS) to survive in host cells and to maintain chronic infection. By as-yet-unknown pathways, the T4SS also elicits inflammatory responses in infected cells. Here we show that inflammation caused by the T4SS results in part from the sensing of a T4SS substrate, VceC, that localizes to the endoplasmic reticulum (ER), an intracellular site of Brucella replication. Possibly via binding of the ER chaperone BiP, VceC causes ER stress with concomitant expression of proinflammatory cytokines. Thus, induction of the unfolded protein response may represent a novel pathway by which host cells can detect pathogens deploying a T4SS.


Subject(s)
Bacterial Secretion Systems , Brucella abortus/metabolism , Brucella abortus/pathogenicity , Unfolded Protein Response , Virulence Factors/metabolism , Animals , Cytokines/metabolism , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/ultrastructure , Endoplasmic Reticulum Chaperone BiP , Female , HeLa Cells , Humans , Inflammation Mediators/metabolism , Macrophages , Mice , Mice, Inbred C57BL , Virulence Factors/toxicity
18.
Cell Host Microbe ; 11(1): 33-45, 2012 Jan 19.
Article in English | MEDLINE | ID: mdl-22264511

ABSTRACT

Autophagy is a cellular degradation process that can capture and eliminate intracellular microbes by delivering them to lysosomes for destruction. However, pathogens have evolved mechanisms to subvert this process. The intracellular bacterium Brucella abortus ensures its survival by forming the Brucella-containing vacuole (BCV), which traffics from the endocytic compartment to the endoplasmic reticulum (ER), where the bacterium proliferates. We show that Brucella replication in the ER is followed by BCV conversion into a compartment with autophagic features (aBCV). While Brucella trafficking to the ER was unaffected in autophagy-deficient cells, aBCV formation required the autophagy-initiation proteins ULK1, Beclin 1, and ATG14L and PI3-kinase activity. However, aBCV formation was independent of the autophagy-elongation proteins ATG5, ATG16L1, ATG4B, ATG7, and LC3B. Furthermore, aBCVs were required to complete the intracellular Brucella lifecycle and for cell-to-cell spreading, demonstrating that Brucella selectively co-opts autophagy-initiation complexes to subvert host clearance and promote infection.


Subject(s)
Autophagy/immunology , Brucella abortus/immunology , Brucella abortus/pathogenicity , Immune Evasion , Vacuoles/microbiology , Animals , Brucella abortus/growth & development , Cell Line , Endocytosis , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/microbiology , Humans , Mice , Mice, Knockout , Protein Transport , Vacuoles/metabolism
19.
Traffic ; 11(5): 626-36, 2010 May.
Article in English | MEDLINE | ID: mdl-20163571

ABSTRACT

We used multiple approaches to investigate the coordination of trans and medial Rab proteins in the regulation of intra-Golgi retrograde trafficking. We reasoned that medially located Rab33b might act downstream of the trans Golgi Rab, Rab6, in regulating intra-Golgi retrograde trafficking. We found that knockdown of Rab33b, like Rab6, suppressed conserved oligomeric Golgi (COG) complex- or Zeste White 10 (ZW10)-depletion induced disruption of the Golgi ribbon in HeLa cells. Moreover, efficient GTP-restricted Rab6 induced relocation of Golgi enzymes to the endoplasmic reticulum (ER) was Rab33b-dependent, but not vice versa, suggesting that the two Rabs act sequentially in an intra-Golgi Rab cascade. In support of this hypothesis, we found that overexpression of GTP-Rab33b induced the dissociation of Rab6 from Golgi membranes in vivo. In addition, the transport of Shiga-like toxin B fragment (SLTB) from the trans to cis Golgi and ER required Rab33b. Surprisingly, depletion of Rab33b had little, if any, immediate effect on cell growth and multiplication. Furthermore, anterograde trafficking of tsO45G protein through the Golgi apparatus was normal. We suggest that the Rab33b/Rab6 regulated intra-Golgi retrograde trafficking pathway must coexist with other Golgi trafficking pathways. In conclusion, we provide the first evidence that Rab33b and Rab6 act to coordinate a major intra-Golgi retrograde trafficking pathway. This coordination may have parallels with Rab conversion/cascade events that regulate endosome, phagosome and exocytic processes.


Subject(s)
Golgi Apparatus/metabolism , Homeostasis/genetics , Biological Transport/genetics , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Golgi Apparatus/drug effects , Golgi Apparatus/genetics , Guanosine Triphosphate/genetics , Guanosine Triphosphate/metabolism , Guanosine Triphosphate/pharmacology , HeLa Cells , Humans , Protein Transport/genetics , Shiga Toxins/genetics , Shiga Toxins/metabolism , Shiga Toxins/pharmacology
20.
Methods Mol Biol ; 457: 179-92, 2008.
Article in English | MEDLINE | ID: mdl-19066027

ABSTRACT

With current light microscopy and laboratory-level computational capability, many questions in organelle assembly and membrane trafficking that were once treated in a qualitative manner can now be treated quantitatively. We present here an overview of the principles involved in doing quantitative fluorescence microscopy. We illustrate these with examples drawn from our work with the Golgi apparatus and endosomes in cultured mammalian cells. The principles themselves can be applied to any system.


Subject(s)
Cell Membrane/metabolism , Microscopy, Fluorescence/methods , Organelles/metabolism , Animals , Biological Transport , Diffusion , Fluorescence Recovery After Photobleaching , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...