Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Dis Model Mech ; 13(7)2020 07 20.
Article in English | MEDLINE | ID: mdl-32461265

ABSTRACT

Toxoplasma gondii is an obligate intracellular parasite capable of invading any nucleated cell. Three main clonal lineages (type I, II, III) exist and murine models have driven the understanding of general and strain-specific immune mechanisms underlying Toxoplasma infection. However, murine models are limited for studying parasite-leukocyte interactions in vivo, and discrepancies exist between cellular immune responses observed in mouse versus human cells. Here, we developed a zebrafish infection model to study the innate immune response to Toxoplasma in vivo By infecting the zebrafish hindbrain ventricle, and using high-resolution microscopy techniques coupled with computer vision-driven automated image analysis, we reveal that Toxoplasma invades brain cells and replicates inside a parasitophorous vacuole to which type I and III parasites recruit host cell mitochondria. We also show that type II and III strains maintain a higher infectious burden than type I strains. To understand how parasites are cleared in vivo, we further analyzed Toxoplasma-macrophage interactions using time-lapse microscopy and three-dimensional correlative light and electron microscopy (3D CLEM). Time-lapse microscopy revealed that macrophages are recruited to the infection site and play a key role in Toxoplasma control. High-resolution 3D CLEM revealed parasitophorous vacuole breakage in brain cells and macrophages in vivo, suggesting that cell-intrinsic mechanisms may be used to destroy the intracellular niche of tachyzoites. Together, our results demonstrate in vivo control of Toxoplasma by macrophages, and highlight the possibility that zebrafish may be further exploited as a novel model system for discoveries within the field of parasite immunity.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Macrophages/parasitology , Rhombencephalon/microbiology , Toxoplasma/growth & development , Toxoplasmosis, Animal/parasitology , Toxoplasmosis, Cerebral/parasitology , Zebrafish/parasitology , Animals , Disease Models, Animal , Host-Parasite Interactions , Macrophages/immunology , Macrophages/ultrastructure , Microscopy, Confocal , Microscopy, Electron, Scanning , Microscopy, Fluorescence , Microscopy, Video , Parasite Load , Rhombencephalon/immunology , Rhombencephalon/ultrastructure , Time Factors , Toxoplasma/immunology , Toxoplasma/ultrastructure , Toxoplasmosis, Animal/immunology , Toxoplasmosis, Animal/pathology , Toxoplasmosis, Cerebral/immunology , Toxoplasmosis, Cerebral/pathology
2.
PLoS Pathog ; 13(6): e1006467, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28650995

ABSTRACT

Shigella flexneri, a Gram-negative enteroinvasive pathogen, causes inflammatory destruction of the human intestinal epithelium. Infection by S. flexneri has been well-studied in vitro and is a paradigm for bacterial interactions with the host immune system. Recent work has revealed that components of the cytoskeleton have important functions in innate immunity and inflammation control. Septins, highly conserved cytoskeletal proteins, have emerged as key players in innate immunity to bacterial infection, yet septin function in vivo is poorly understood. Here, we use S. flexneri infection of zebrafish (Danio rerio) larvae to study in vivo the role of septins in inflammation and infection control. We found that depletion of Sept15 or Sept7b, zebrafish orthologs of human SEPT7, significantly increased host susceptibility to bacterial infection. Live-cell imaging of Sept15-depleted larvae revealed increasing bacterial burdens and a failure of neutrophils to control infection. Strikingly, Sept15-depleted larvae present significantly increased activity of Caspase-1 and more cell death upon S. flexneri infection. Dampening of the inflammatory response with anakinra, an antagonist of interleukin-1 receptor (IL-1R), counteracts Sept15 deficiency in vivo by protecting zebrafish from hyper-inflammation and S. flexneri infection. These findings highlight a new role for septins in host defence against bacterial infection, and suggest that septin dysfunction may be an underlying factor in cases of hyper-inflammation.


Subject(s)
Dysentery, Bacillary/immunology , Immunity, Innate/immunology , Septins/metabolism , Animals , Disease Models, Animal , Dysentery, Bacillary/microbiology , Host-Pathogen Interactions/immunology , Humans , Inflammation/immunology , Inflammation/microbiology , Intestinal Mucosa/microbiology , Larva/metabolism , Neutrophils/metabolism , Neutrophils/microbiology , Shigella flexneri , Zebrafish
3.
Nat Commun ; 8: 15048, 2017 04 20.
Article in English | MEDLINE | ID: mdl-28425465

ABSTRACT

Aquatic chytrid fungi threaten amphibian biodiversity worldwide owing to their ability to rapidly expand their geographical distributions and to infect a wide range of hosts. Combating this risk requires an understanding of chytrid host range to identify potential reservoirs of infection and to safeguard uninfected regions through enhanced biosecurity. Here we extend our knowledge on the host range of the chytrid Batrachochytrium dendrobatidis by demonstrating infection of a non-amphibian vertebrate host, the zebrafish. We observe dose-dependent mortality and show that chytrid can infect and proliferate on zebrafish tissue. We also show that infection phenotypes (fin erosion, cell apoptosis and muscle degeneration) are direct symptoms of infection. Successful infection is dependent on disrupting the zebrafish microbiome, highlighting that, as is widely found in amphibians, commensal bacteria confer protection against this pathogen. Collectively, our findings greatly expand the limited tool kit available to study pathogenesis and host response to chytrid infection.


Subject(s)
Amphibians/microbiology , Chytridiomycota/physiology , Fish Diseases/microbiology , Zebrafish/microbiology , Animal Fins/microbiology , Animals , Host-Pathogen Interactions , Microbial Interactions , Microbiota/physiology
4.
Nat Microbiol ; 2: 17027, 2017 Mar 06.
Article in English | MEDLINE | ID: mdl-28263305

ABSTRACT

Pseudomonas aeruginosa is a Gram-negative bacterial pathogen associated with acute and chronic infections. The universal cyclic-di-GMP second messenger is instrumental in the switch from a motile lifestyle to resilient biofilm as in the cystic fibrosis lung. The SadC diguanylate cyclase is associated with this patho-adaptive transition. Here, we identify an unrecognized SadC partner, WarA, which we show is a methyltransferase in complex with a putative kinase, WarB. We established that WarA binds to cyclic-di-GMP, which potentiates its methyltransferase activity. Together, WarA and WarB have structural similarities with the bifunctional Escherichia coli lipopolysaccharide (LPS) O antigen regulator WbdD. Strikingly, WarA influences P. aeruginosa O antigen modal distribution and interacts with the LPS biogenesis machinery. LPS is known to modulate the immune response in the host, and by using a zebrafish infection model, we implicate WarA in the ability of P. aeruginosa to evade detection by the host.


Subject(s)
Cyclic GMP/analogs & derivatives , Immune Evasion , Lipopolysaccharides/metabolism , Pseudomonas aeruginosa/metabolism , Pseudomonas aeruginosa/pathogenicity , Animals , Cyclic GMP/metabolism , Disease Models, Animal , Methyltransferases/metabolism , Protein Binding , Pseudomonas Infections/microbiology , Pseudomonas Infections/pathology , Zebrafish
5.
EMBO Rep ; 18(2): 303-318, 2017 02.
Article in English | MEDLINE | ID: mdl-28039206

ABSTRACT

During infection, plasma membrane (PM) blebs protect host cells against bacterial pore-forming toxins (PFTs), but were also proposed to promote pathogen dissemination. However, the details and impact of blebbing regulation during infection remained unclear. Here, we identify the endoplasmic reticulum chaperone Gp96 as a novel regulator of PFT-induced blebbing. Gp96 interacts with non-muscle myosin heavy chain IIA (NMHCIIA) and controls its activity and remodelling, which is required for appropriate coordination of bleb formation and retraction. This mechanism involves NMHCIIA-Gp96 interaction and their recruitment to PM blebs and strongly resembles retraction of uropod-like structures from polarized migrating cells, a process that also promotes NMHCIIA-Gp96 association. Consistently, Gp96 and NMHCIIA not only protect the PM integrity from listeriolysin O (LLO) during infection by Listeria monocytogenes but also affect cytoskeletal organization and cell migration. Finally, we validate the association between Gp96 and NMHCIIA in vivo and show that Gp96 is required to protect hosts from LLO-dependent killing.


Subject(s)
Actomyosin/metabolism , Bacterial Toxins/metabolism , Membrane Glycoproteins/metabolism , Pore Forming Cytotoxic Proteins/metabolism , Animals , Bacterial Proteins/metabolism , Cell Survival , Humans , Listeria monocytogenes , Mice , Molecular Chaperones/metabolism , Zebrafish
6.
J Vis Exp ; (91): e51601, 2014 Sep 09.
Article in English | MEDLINE | ID: mdl-25226510

ABSTRACT

Shigella flexneri is an intracellular pathogen that can escape from phagosomes to reach the cytosol, and polymerize the host actin cytoskeleton to promote its motility and dissemination. New work has shown that proteins involved in actin-based motility are also linked to autophagy, an intracellular degradation process crucial for cell autonomous immunity. Strikingly, host cells may prevent actin-based motility of S. flexneri by compartmentalizing bacteria inside 'septin cages' and targeting them to autophagy. These observations indicate that a more complete understanding of septins, a family of filamentous GTP-binding proteins, will provide new insights into the process of autophagy. This report describes protocols to monitor autophagy-cytoskeleton interactions caused by S. flexneri in vitro using tissue culture cells and in vivo using zebrafish larvae. These protocols enable investigation of intracellular mechanisms that control bacterial dissemination at the molecular, cellular, and whole organism level.


Subject(s)
Dysentery, Bacillary/microbiology , Shigella flexneri/cytology , Animals , Autophagy/physiology , Cytoskeleton/microbiology , Cytoskeleton/pathology , Dysentery, Bacillary/pathology , Female , Male , Shigella flexneri/pathogenicity , Zebrafish
7.
PLoS Pathog ; 9(9): e1003588, 2013.
Article in English | MEDLINE | ID: mdl-24039575

ABSTRACT

Autophagy, an ancient and highly conserved intracellular degradation process, is viewed as a critical component of innate immunity because of its ability to deliver cytosolic bacteria to the lysosome. However, the role of bacterial autophagy in vivo remains poorly understood. The zebrafish (Danio rerio) has emerged as a vertebrate model for the study of infections because it is optically accessible at the larval stages when the innate immune system is already functional. Here, we have characterized the susceptibility of zebrafish larvae to Shigella flexneri, a paradigm for bacterial autophagy, and have used this model to study Shigella-phagocyte interactions in vivo. Depending on the dose, S. flexneri injected in zebrafish larvae were either cleared in a few days or resulted in a progressive and ultimately fatal infection. Using high resolution live imaging, we found that S. flexneri were rapidly engulfed by macrophages and neutrophils; moreover we discovered a scavenger role for neutrophils in eliminating infected dead macrophages and non-immune cell types that failed to control Shigella infection. We observed that intracellular S. flexneri could escape to the cytosol, induce septin caging and be targeted to autophagy in vivo. Depletion of p62 (sequestosome 1 or SQSTM1), an adaptor protein critical for bacterial autophagy in vitro, significantly increased bacterial burden and host susceptibility to infection. These results show the zebrafish larva as a new model for the study of S. flexneri interaction with phagocytes, and the manipulation of autophagy for anti-bacterial therapy in vivo.


Subject(s)
Autophagy , Dysentery, Bacillary/metabolism , Macrophages/metabolism , Neutrophils/metabolism , Shigella flexneri/metabolism , Zebrafish/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Disease Models, Animal , Dysentery, Bacillary/genetics , Dysentery, Bacillary/pathology , Humans , Macrophages/microbiology , Macrophages/pathology , Neutrophils/microbiology , Neutrophils/pathology , Zebrafish/genetics , Zebrafish/microbiology , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...