Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Front Cell Infect Microbiol ; 10: 596023, 2020.
Article in English | MEDLINE | ID: mdl-33585270

ABSTRACT

Invasive infections due to group A Streptococcus (GAS) advance rapidly causing tissue degradation and unregulated inflammation. Neutrophils are the primary immune cells that respond to GAS. The neutrophil response to GAS was characterised in response to two M1T1 isolates; 5448 and animal passaged variant 5448AP. Co-incubation of neutrophils with 5448AP resulted in proliferation of GAS and lowered the production of reactive oxygen species when compared with 5448. Infection with both strains invoked neutrophil death, however apoptosis was reduced in response to 5448AP. Both strains induced neutrophil caspase-1 and caspase-4 expression in vitro, with inflammatory caspase activation detected in vitro and in vivo. GAS infections involving strains such as 5448AP that promote an inflammatory neutrophil phenotype may contribute to increased inflammation yet ineffective bacterial eradication, contributing to the severity of invasive GAS infections.


Subject(s)
Streptococcal Infections , Streptococcus pyogenes , Animals , Caspases/genetics , Neutrophils , Phenotype
2.
Trends Microbiol ; 26(5): 401-410, 2018 05.
Article in English | MEDLINE | ID: mdl-29548832

ABSTRACT

Both extracellular RNAs and extracellular vesicles (EVs) have recently garnered attention as novel mediators of intercellular communication in eukaryotes and prokaryotes alike. EVs not only permit export of RNA, but also facilitate delivery and trans-kingdom exchange of these and other biomolecules, for instance between microbes and their hosts. In this Opinion article, we propose that EV-mediated export of RNA represents a universal mechanism for interkingdom and intrakingdom communication that is conserved among bacterial, archaeal, and eukaryotic microbes. We speculate how microbes might use EV RNA to influence target cell gene expression or manipulate host immune responses.


Subject(s)
Cell Communication/physiology , Extracellular Vesicles/metabolism , RNA/metabolism , Archaea/physiology , Eukaryota/physiology , Gene Expression , Gene Silencing , Immunity, Innate , Prokaryotic Cells/physiology
3.
J Innate Immun ; 7(6): 612-22, 2015.
Article in English | MEDLINE | ID: mdl-25997401

ABSTRACT

Polymorphonuclear leukocyte (PMN) cell death strongly influences the resolution of inflammatory episodes, and may exacerbate adverse pathologies in response to infection. We investigated PMN cell death mechanisms following infection by virulent group A Streptococcus (GAS). Human PMNs were infected in vitro with a clinical, virulent GAS isolate and an avirulent derivative strain, and compared for phagocytosis, the production of reactive oxygen species (ROS), mitochondrial membrane depolarization and apoptotic markers. C57BL/6J mice were then infected, in order to observe the effects on murine PMNs in vivo. Human PMNs phagocytosed virulent GAS less efficiently, produced less ROS and underwent reduced mitochondrial membrane depolarization compared with phagocytosis of avirulent GAS. Morphological and biochemical analyses revealed that PMNs infected with avirulent GAS exhibited nuclear fragmentation and caspase-3 activation consistent with an anti-inflammatory apoptotic phenotype. Conversely, virulent GAS induced PMN vacuolization and plasma membrane permeabilization, leading to a necrotic form of cell death. Infection of the mice with virulent GAS engendered significantly higher systemic pro-inflammatory cytokine release and localized infiltration of murine PMNs, with cells associated with virulent GAS infection exhibiting reduced apoptotic potential. Avirulent GAS infection was associated with lower levels of proinflammatory cytokines and tissue PMN apoptosis. We propose that the differences in PMN cell death mechanisms influence the inflammatory responses to infection by GAS.


Subject(s)
Apoptosis/immunology , Caspase 3/immunology , Neutrophils/immunology , Phagocytosis , Streptococcal Infections/immunology , Streptococcus pyogenes/immunology , Animals , Female , Humans , Male , Mice , Mitochondrial Membranes/immunology , Mitochondrial Membranes/pathology , Necrosis , Reactive Oxygen Species/immunology , Streptococcal Infections/pathology
4.
PLoS Pathog ; 10(8): e1004266, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25165887

ABSTRACT

Infections caused by group A Streptococcus (GAS) are characterized by robust inflammatory responses and can rapidly lead to life-threatening disease manifestations. However, host mechanisms that respond to GAS, which may influence disease pathology, are understudied. Recent works indicate that GAS infection is recognized by multiple extracellular and intracellular receptors and activates cell signalling via discrete pathways. Host leukocyte receptor binding to GAS-derived products mediates release of inflammatory mediators associated with severe GAS disease. GAS induces divergent phagocyte programmed cell death responses and has inflammatory implications. Epithelial cell apoptotic and autophagic components are mobilized by GAS infection, but can be subverted to ensure bacterial survival. Examination of host interactions with GAS and consequences of GAS infection in the context of cellular receptors responsible for GAS recognition, inflammatory mediator responses, and cell death mechanisms, highlights potential avenues for diagnostic and therapeutic intervention. Understanding the molecular and cellular basis of host symptoms during severe GAS disease will assist the development of improved treatment regimens for this formidable pathogen.


Subject(s)
Host-Parasite Interactions/physiology , Inflammation/microbiology , Streptococcal Infections/microbiology , Animals , Cell Death/physiology , Humans , Inflammation/immunology , Inflammation/physiopathology , Streptococcal Infections/immunology , Streptococcal Infections/physiopathology , Streptococcus pyogenes
5.
J Innate Immun ; 6(2): 240-50, 2014.
Article in English | MEDLINE | ID: mdl-23969887

ABSTRACT

The globally significant human pathogen group A Streptococcus (GAS) sequesters the host protease plasmin to the cell surface during invasive disease initiation. Recent evidence has shown that localized plasmin activity prevents opsonization of several bacterial species by key components of the innate immune system in vitro. Here we demonstrate that plasmin at the GAS cell surface resulted in degradation of complement factor C3b, and that plasminogen acquisition is associated with a decrease in C3b opsonization and neutrophil-mediated killing in vitro. Furthermore, the ability to acquire cell surface plasmin(ogen) correlates directly with a decrease in C3b opsonization, neutrophil phagocytosis, and increased bacterial survival in a humanized plasminogen mouse model of infection. These findings demonstrate that localized plasmin(ogen) plays an important role in facilitating GAS escape from the host innate immune response and increases bacterial virulence in the early stages of infection.


Subject(s)
Complement C3b/immunology , Neutrophils/immunology , Phagocytosis/immunology , Plasminogen/immunology , Streptococcus pyogenes/immunology , Animals , Blotting, Western , Complement C3b/metabolism , Female , Fibrinolysin/immunology , Fibrinolysin/metabolism , Flow Cytometry , Host-Pathogen Interactions/immunology , Humans , Immune Evasion/immunology , Male , Mice, Transgenic , Neutrophils/metabolism , Neutrophils/microbiology , Plasminogen/genetics , Plasminogen/metabolism , Streptococcal Infections/genetics , Streptococcal Infections/immunology , Streptococcal Infections/microbiology , Streptococcus pyogenes/metabolism , Streptococcus pyogenes/physiology , Streptokinase/immunology , Streptokinase/metabolism
6.
FASEB J ; 27(7): 2633-43, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23531597

ABSTRACT

In Western countries, invasive infections caused by M1T1 serotype group A Streptococcus (GAS) are epidemiologically linked to mutations in the control of virulence regulatory 2-component operon (covRS). In indigenous communities and developing countries, severe GAS disease is associated with genetically diverse non-M1T1 GAS serotypes. Hypervirulent M1T1 covRS mutant strains arise through selection by human polymorphonuclear cells for increased expression of GAS virulence factors such as the DNase Sda1, which promotes neutrophil resistance. The GAS bacteremia isolate NS88.2 (emm 98.1) is a covS mutant that exhibits a hypervirulent phenotype and neutrophil resistance yet lacks the phage-encoded Sda1. Here, we have employed a comprehensive systems biology (genomic, transcriptomic, and proteomic) approach to identify NS88.2 virulence determinants that enhance neutrophil resistance in the non-M1T1 GAS genetic background. Using this approach, we have identified streptococcal collagen-like protein A and general stress protein 24 proteins as NS88.2 determinants that contribute to survival in whole blood and neutrophil resistance in non-M1T1 GAS. This study has revealed new factors that contribute to GAS pathogenicity that may play important roles in resisting innate immune defenses and the development of human invasive infections.


Subject(s)
Bacterial Proteins/immunology , Streptococcal Infections/immunology , Streptococcus pyogenes/immunology , Animals , Bacterial Adhesion/genetics , Bacterial Adhesion/immunology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Line , Electrophoresis, Gel, Two-Dimensional , Gene Expression Profiling , Gene Expression Regulation, Bacterial , Genome, Bacterial/genetics , Genomics/methods , Host-Pathogen Interactions/immunology , Humans , Mice , Microbial Viability/genetics , Microbial Viability/immunology , Mutation , Neutrophil Activation/immunology , Neutrophils/immunology , Neutrophils/metabolism , Neutrophils/microbiology , Oligonucleotide Array Sequence Analysis , Proteome/genetics , Proteome/metabolism , Proteomics/methods , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Streptococcal Infections/blood , Streptococcal Infections/microbiology , Streptococcus pyogenes/genetics , Streptococcus pyogenes/pathogenicity , Virulence/genetics , Virulence/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...