Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters











Database
Language
Publication year range
1.
Autophagy Rep ; 2(1)2023.
Article in English | MEDLINE | ID: mdl-37325703

ABSTRACT

Septins are cytoskeletal proteins implicated in numerous cellular processes including cytokinesis and morphogenesis. In the case of infection by Shigella flexneri, septins assemble into cage-like structures that entrap cytosolic bacteria targeted by autophagy. The interplay between septin cage entrapment and bacterial autophagy is poorly understood. We used a correlative light and cryo-soft X-ray tomography (cryo-SXT) pipeline to study septin cage entrapment of Shigella in its near-native state. Septin cages could be identified as X-ray dense structures, indicating they contain host cell proteins and lipids consistent with their autophagy links. Airyscan confocal microscopy of Shigella-septin cages showed that septins and lysine 63 (K63)-linked ubiquitin chains are present in separate bacterial microdomains, suggesting they are recruited separately. Finally, Cryo-SXT and live-cell imaging revealed an interaction between septins and microtubule-associated protein light chain 3B (LC3B)-positive membranes during autophagy of Shigella. Collectively our data present a new model for how septin-caged Shigella are targeted to autophagy.

2.
mBio ; 14(4): e0088223, 2023 08 31.
Article in English | MEDLINE | ID: mdl-37255304

ABSTRACT

Enteroinvasive Escherichia coli (EIEC) and Shigella are closely related agents of bacillary dysentery. It is widely viewed that EIEC and Shigella species evolved from E. coli via independent acquisitions of a large virulence plasmid (pINV) encoding a type 3 secretion system (T3SS). Sequence Type (ST)99 O96:H19 E. coli is a novel clone of EIEC responsible for recent outbreaks in Europe and South America. Here, we use 92 whole genome sequences to reconstruct a dated phylogeny of ST99 E. coli, revealing distinct phylogenomic clusters of pINV-positive and -negative isolates. To study the impact of pINV acquisition on the virulence of this clone, we developed an EIEC-zebrafish infection model showing that virulence of ST99 EIEC is thermoregulated. Strikingly, zebrafish infection using a T3SS-deficient ST99 EIEC strain and the oldest available pINV-negative isolate reveals a separate, temperature-independent mechanism of virulence, indicating that ST99 non-EIEC strains were virulent before pINV acquisition. Taken together, these results suggest that an already pathogenic E. coli acquired pINV and that virulence of ST99 isolates became thermoregulated once pINV was acquired. IMPORTANCE Enteroinvasive Escherichia coli (EIEC) and Shigella are etiological agents of bacillary dysentery. Sequence Type (ST)99 is a clone of EIEC hypothesized to cause human disease by the recent acquisition of pINV, a large plasmid encoding a type 3 secretion system (T3SS) that confers the ability to invade human cells. Using Bayesian analysis and zebrafish larvae infection, we show that the virulence of ST99 EIEC isolates is highly dependent on temperature, while T3SS-deficient isolates encode a separate temperature-independent mechanism of virulence. These results indicate that ST99 non-EIEC isolates may have been virulent before pINV acquisition and highlight an important role of pINV acquisition in the dispersal of ST99 EIEC in humans, allowing wider dissemination across Europe and South America.


Subject(s)
Dysentery, Bacillary , Escherichia coli Infections , Shigella , Animals , Humans , Escherichia coli , Virulence/genetics , Zebrafish , Type III Secretion Systems/genetics , Bayes Theorem , Temperature , Plasmids/genetics , Shigella/genetics
3.
Dis Model Mech ; 16(6)2023 06 01.
Article in English | MEDLINE | ID: mdl-37161932

ABSTRACT

Shigella are Gram-negative bacterial pathogens responsible for bacillary dysentery (also called shigellosis). The absence of a licensed vaccine and widespread emergence of antibiotic resistance has led the World Health Organisation (WHO) to highlight Shigella as a priority pathogen requiring urgent attention. Several infection models have been useful to explore the Shigella infection process; yet, we still lack information regarding events taking place in vivo. Here, using a Shigella-zebrafish infection model and high-content microscopy, we developed an automated microscopy workflow to non-invasively study fluorescently labelled bacteria and neutrophils in vivo. We applied our workflow to antibiotic-treated zebrafish, and demonstrate that antibiotics reduce bacterial burden and not neutrophil recruitment to the hindbrain ventricle. We discovered that nalidixic acid (a bactericidal antibiotic) can work with leukocytes in an additive manner to control Shigella flexneri infection and can also restrict dissemination of Shigella sonnei from the hindbrain ventricle. We envision that our automated microscopy workflow, applied here to study the interactions between Shigella and neutrophils as well as antibiotic efficacy in zebrafish, can be useful to innovate treatments for infection control in humans.


Subject(s)
Dysentery, Bacillary , Shigella , Humans , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Zebrafish , Microscopy , Workflow , Dysentery, Bacillary/drug therapy
4.
J Cell Sci ; 136(7)2023 04 01.
Article in English | MEDLINE | ID: mdl-36939083

ABSTRACT

During host cell invasion, Shigella escapes to the cytosol and polymerizes actin for cell-to-cell spread. To restrict cell-to-cell spread, host cells employ cell-autonomous immune responses including antibacterial autophagy and septin cage entrapment. How septins interact with the autophagy process to target Shigella for destruction is poorly understood. Here, we employed a correlative light and cryo-soft X-ray tomography (cryo-SXT) pipeline to study Shigella septin cage entrapment in its near-native state. Quantitative cryo-SXT showed that Shigella fragments mitochondria and enabled visualization of X-ray-dense structures (∼30 nm resolution) surrounding Shigella entrapped in septin cages. Using Airyscan confocal microscopy, we observed lysine 63 (K63)-linked ubiquitin chains decorating septin-cage-entrapped Shigella. Remarkably, septins and K63 chains are present in separate bacterial microdomains, indicating they are recruited separately during antibacterial autophagy. Cryo-SXT and live-cell imaging revealed an interaction between septins and LC3B-positive membranes during autophagy of Shigella. Together, these findings demonstrate how septin-caged Shigella are targeted for autophagy and provide fundamental insights into autophagy-cytoskeleton interactions.


Subject(s)
Septins , Shigella , Septins/metabolism , Shigella/metabolism , Cytoskeleton/metabolism , Autophagy/physiology , Ubiquitins/metabolism
5.
Nat Commun ; 12(1): 6764, 2021 11 19.
Article in English | MEDLINE | ID: mdl-34799563

ABSTRACT

The field of cellular microbiology, rooted in the co-evolution of microbes and their hosts, studies intracellular pathogens and their manipulation of host cell machinery. In this review, we highlight emerging technologies and infection models that recently promoted opportunities in cellular microbiology. We overview the explosion of microscopy techniques and how they reveal unprecedented detail at the host-pathogen interface. We discuss the incorporation of robotics and artificial intelligence to image-based screening modalities, biochemical mapping approaches, as well as dual RNA-sequencing techniques. Finally, we describe chips, organoids and animal models used to dissect biophysical and in vivo aspects of the infection process. As our knowledge of the infected cell improves, cellular microbiology holds great promise for development of anti-infective strategies with translational applications in human health.


Subject(s)
Anti-Infective Agents/pharmacology , Cytological Techniques , Infections/drug therapy , Microbiological Techniques , Microscopy/methods , Animals , Anti-Infective Agents/therapeutic use , Artificial Intelligence , Disease Models, Animal , Drug Evaluation, Preclinical/methods , Drug Evaluation, Preclinical/trends , Host-Pathogen Interactions , Humans , Infections/microbiology , Microscopy/trends , Organoids , Sequence Analysis, RNA , Translational Research, Biomedical/methods , Translational Research, Biomedical/trends
6.
PLoS Pathog ; 13(4): e1006344, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28414774

ABSTRACT

Autophagy is a eukaryotic catabolic process also participating in cell-autonomous defence. Infected host cells generate double-membrane autophagosomes that mature in autolysosomes to engulf, kill and digest cytoplasmic pathogens. However, several bacteria subvert autophagy and benefit from its machinery and functions. Monitoring infection stages by genetics, pharmacology and microscopy, we demonstrate that the ESX-1 secretion system of Mycobacterium marinum, a close relative to M. tuberculosis, upregulates the transcription of autophagy genes, and stimulates autophagosome formation and recruitment to the mycobacteria-containing vacuole (MCV) in the host model organism Dictyostelium. Antagonistically, ESX-1 is also essential to block the autophagic flux and deplete the MCV of proteolytic activity. Activators of the TORC1 complex localize to the MCV in an ESX-1-dependent manner, suggesting an important role in the manipulation of autophagy by mycobacteria. Our findings suggest that the infection by M. marinum activates an autophagic response that is simultaneously repressed and exploited by the bacterium to support its survival inside the MCV.


Subject(s)
Autophagy , Bacterial Proteins/metabolism , Multiprotein Complexes/metabolism , Mycobacterium Infections, Nontuberculous/metabolism , Mycobacterium Infections, Nontuberculous/physiopathology , Mycobacterium marinum/metabolism , TOR Serine-Threonine Kinases/metabolism , Bacterial Proteins/genetics , Dictyostelium/genetics , Dictyostelium/metabolism , Dictyostelium/microbiology , Host-Pathogen Interactions , Humans , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes/genetics , Mycobacterium Infections, Nontuberculous/genetics , Mycobacterium Infections, Nontuberculous/virology , Mycobacterium marinum/genetics , TOR Serine-Threonine Kinases/genetics , Vacuoles/microbiology
7.
Front Immunol ; 8: 1906, 2017.
Article in English | MEDLINE | ID: mdl-29354124

ABSTRACT

The soil-dwelling social amoeba Dictyostelium discoideum feeds on bacteria. Each meal is a potential infection because some bacteria have evolved mechanisms to resist predation. To survive such a hostile environment, D. discoideum has in turn evolved efficient antimicrobial responses that are intertwined with phagocytosis and autophagy, its nutrient acquisition pathways. The core machinery and antimicrobial functions of these pathways are conserved in the mononuclear phagocytes of mammals, which mediate the initial, innate-immune response to infection. In this review, we discuss the advantages and relevance of D. discoideum as a model phagocyte to study cell-autonomous defenses. We cover the antimicrobial functions of phagocytosis and autophagy and describe the processes that create a microbicidal phagosome: acidification and delivery of lytic enzymes, generation of reactive oxygen species, and the regulation of Zn2+, Cu2+, and Fe2+ availability. High concentrations of metals poison microbes while metal sequestration inhibits their metabolic activity. We also describe microbial interference with these defenses and highlight observations made first in D. discoideum. Finally, we discuss galectins, TNF receptor-associated factors, tripartite motif-containing proteins, and signal transducers and activators of transcription, microbial restriction factors initially characterized in mammalian phagocytes that have either homologs or functional analogs in D. discoideum.

8.
Lab Chip ; 16(17): 3276-85, 2016 08 16.
Article in English | MEDLINE | ID: mdl-27425421

ABSTRACT

The impact of cellular individuality on host-microbe interactions is increasingly appreciated but studying the temporal dynamics of single-cell behavior in this context remains technically challenging. Here we present a microfluidic platform, InfectChip, to trap motile infected cells for high-resolution time-lapse microscopy. This approach allows the direct visualization of all stages of infection, from bacterial uptake to death of the bacterium or host cell, over extended periods of time. We demonstrate the utility of this approach by co-culturing an established host-cell model, Dictyostelium discoideum, with the extracellular pathogen Klebsiella pneumoniae or the intracellular pathogen Mycobacterium marinum. We show that the outcome of such infections is surprisingly heterogeneous, ranging from abortive infection to death of the bacterium or host cell. InfectChip thus provides a simple method to dissect the time-course of host-microbe interactions at the single-cell level, yielding new insights that could not be gleaned from conventional population-based measurements.


Subject(s)
Cell Tracking/instrumentation , Coculture Techniques/instrumentation , Host-Pathogen Interactions , Lab-On-A-Chip Devices , Models, Biological , Phagocytosis , Single-Cell Analysis/instrumentation , Animals , Cells, Cultured , Cells, Immobilized , Computer-Aided Design , Dictyostelium/cytology , Dictyostelium/immunology , Dictyostelium/physiology , Dictyostelium/ultrastructure , Dimethylpolysiloxanes/chemistry , Equipment Design , Humans , Image Interpretation, Computer-Assisted , Klebsiella pneumoniae/cytology , Klebsiella pneumoniae/immunology , Klebsiella pneumoniae/physiology , Klebsiella pneumoniae/ultrastructure , Microscopy, Confocal , Microscopy, Electron, Scanning , Mycobacterium marinum/cytology , Mycobacterium marinum/immunology , Mycobacterium marinum/physiology , Mycobacterium marinum/ultrastructure , Phagocytes/cytology , Phagocytes/immunology , Phagocytes/microbiology , Phagocytes/parasitology , Time-Lapse Imaging
SELECTION OF CITATIONS
SEARCH DETAIL