Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Front Immunol ; 15: 1382638, 2024.
Article in English | MEDLINE | ID: mdl-38715601

ABSTRACT

Recovery from respiratory pneumococcal infections generates lung-localized protection against heterotypic bacteria, mediated by resident memory lymphocytes. Optimal protection in mice requires re-exposure to pneumococcus within days of initial infection. Serial surface marker phenotyping of B cell populations in a model of pneumococcal heterotypic immunity revealed that bacterial re-exposure stimulates the immediate accumulation of dynamic and heterogeneous populations of B cells in the lung, and is essential for the establishment of lung resident memory B (BRM) cells. The B cells in the early wave were activated, proliferating locally, and associated with both CD4+ T cells and CXCL13. Antagonist- and antibody-mediated interventions were implemented during this early timeframe to demonstrate that lymphocyte recirculation, CD4+ cells, and CD40 ligand (CD40L) signaling were all needed for lung BRM cell establishment, whereas CXCL13 signaling was not. While most prominent as aggregates in the loose connective tissue of bronchovascular bundles, morphometry and live lung imaging analyses showed that lung BRM cells were equally numerous as single cells dispersed throughout the alveolar septae. We propose that CD40L signaling from antigen-stimulated CD4+ T cells in the infected lung is critical to establishment of local BRM cells, which subsequently protect the airways and parenchyma against future potential infections.


Subject(s)
CD4-Positive T-Lymphocytes , CD40 Ligand , Lung , Memory B Cells , Streptococcus pneumoniae , Animals , CD40 Ligand/metabolism , CD40 Ligand/immunology , Mice , Streptococcus pneumoniae/immunology , Lung/immunology , Lung/pathology , Lung/microbiology , CD4-Positive T-Lymphocytes/immunology , Memory B Cells/immunology , Memory B Cells/metabolism , Pneumococcal Infections/immunology , Mice, Inbred C57BL , Immunologic Memory , Chemokine CXCL13/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Disease Models, Animal , Signal Transduction , Lymphocyte Activation/immunology
2.
Pneumonia (Nathan) ; 15(1): 4, 2023 Feb 25.
Article in English | MEDLINE | ID: mdl-36829255

ABSTRACT

The lungs are an immunologically unique environment; they are exposed to innumerable pathogens and particulate matter daily. Appropriate clearance of pathogens and response to pollutants is required to prevent overwhelming infection, while preventing tissue damage and maintaining efficient gas exchange. Broadly, the innate immune system is the collection of immediate, intrinsic immune responses to pathogen or tissue injury. In this review, we will examine the innate immune responses of the lung, with a particular focus on their role in pneumonia. We will discuss the anatomic barriers and antimicrobial proteins of the lung, pathogen and injury recognition, and the role of leukocytes (macrophages, neutrophils, and innate lymphocytes) and lung stromal cells in innate immunity. Throughout the review, we will focus on new findings in innate immunity as well as features that are unique to the lung.

3.
JCI Insight ; 7(23)2022 12 08.
Article in English | MEDLINE | ID: mdl-36264633

ABSTRACT

Identifying host factors that contribute to pneumonia incidence and severity are of utmost importance to guiding the development of more effective therapies. Lectin-like oxidized low-density lipoprotein receptor 1 (LOX-1, encoded by OLR1) is a scavenger receptor known to promote vascular injury and inflammation, but whether and how LOX-1 functions in the lung are unknown. Here, we provide evidence of substantial accumulation of LOX-1 in the lungs of patients with acute respiratory distress syndrome and in mice with pneumonia. Unlike previously described injurious contributions of LOX-1, we found that LOX-1 is uniquely protective in the pulmonary airspaces, limiting proteinaceous edema and inflammation. We also identified alveolar macrophages and recruited neutrophils as 2 prominent sites of LOX-1 expression in the lungs, whereby macrophages are capable of further induction during pneumonia and neutrophils exhibit a rapid, but heterogenous, elevation of LOX-1 in the infected lung. Blockade of LOX-1 led to dysregulated immune signaling in alveolar macrophages, marked by alterations in activation markers and a concomitant elevation of inflammatory gene networks. However, bone marrow chimeras also suggested a prominent role for neutrophils in LOX-1-mediated lung protection, further supported by LOX-1+ neutrophils exhibiting transcriptional changes consistent with reparative processes. Taken together, this work establishes LOX-1 as a tissue-protective factor in the lungs during pneumonia, possibly mediated by its influence on immune signaling in alveolar macrophages and LOX-1+ airspace neutrophils.


Subject(s)
Lung Injury , Pneumonia , Scavenger Receptors, Class E , Animals , Mice , Scavenger Receptors, Class E/genetics
4.
Am J Physiol Lung Cell Mol Physiol ; 322(4): L550-L563, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35137631

ABSTRACT

During bacterial pneumonia, alveolar epithelial cells are critical for maintaining gas exchange and providing antimicrobial as well as pro-immune properties. We previously demonstrated that leukemia inhibitory factor (LIF), an IL-6 family cytokine, is produced by type II alveolar epithelial cells (ATII) and is critical for tissue protection during bacterial pneumonia. However, the target cells and mechanisms of LIF-mediated protection remain unknown. Here, we demonstrate that antibody-induced LIF blockade remodels the lung epithelial transcriptome in association with increased apoptosis. Based on these data, we performed pneumonia studies using a novel mouse model in which LIFR (the unique receptor for LIF) is absent in lung epithelium. Although LIFR is expressed on the surface of epithelial cells, its absence only minimally contributed to tissue protection during pneumonia. Single-cell RNA-sequencing (scRNAseq) was conducted to identify adult murine lung cell types most prominently expressing Lifr, revealing endothelial cells, mesenchymal cells, and ATIIs as major sources of Lifr. Sequencing data indicated that ATII cells were significantly impacted by pneumonia, with additional differences observed in response to LIF neutralization, including but not limited to gene programs related to cell death, injury, and inflammation. Overall, our data suggest that LIF signaling on epithelial cells alters responses in this cell type during pneumonia. However, our results also suggest separate and perhaps more prominent roles of LIFR in other cell types, such as endothelial cells or mesenchymal cells, which provide grounds for future investigation.


Subject(s)
Lung Injury , Pneumonia, Bacterial , Animals , Apoptosis , Endothelial Cells/metabolism , Leukemia Inhibitory Factor/genetics , Mice , Signal Transduction
5.
J Immunol ; 207(7): 1891-1902, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34470857

ABSTRACT

Systemic duress, such as that elicited by sepsis, burns, or trauma, predisposes patients to secondary pneumonia, demanding better understanding of host pathways influencing this deleterious connection. These pre-existing circumstances are capable of triggering the hepatic acute-phase response (APR), which we previously demonstrated is essential for limiting susceptibility to secondary lung infections. To identify potential mechanisms underlying protection afforded by the lung-liver axis, our studies aimed to evaluate liver-dependent lung reprogramming when a systemic inflammatory challenge precedes pneumonia. Wild-type mice and APR-deficient littermate mice with hepatocyte-specific deletion of STAT3 (hepSTAT3-/-), a transcription factor necessary for full APR initiation, were challenged i.p. with LPS to induce endotoxemia. After 18 h, pneumonia was induced by intratracheal Escherichia coli instillation. Endotoxemia elicited significant transcriptional alterations in the lungs of wild-type and hepSTAT3-/- mice, with nearly 2000 differentially expressed genes between genotypes. The gene signatures revealed exaggerated immune activity in the lungs of hepSTAT3-/- mice, which were compromised in their capacity to launch additional cytokine responses to secondary infection. Proteomics revealed substantial liver-dependent modifications in the airspaces of pneumonic mice, implicating a network of dispatched liver-derived mediators influencing lung homeostasis. These results indicate that after systemic inflammation, liver acute-phase changes dramatically remodel the lungs, resulting in a modified landscape for any stimuli encountered thereafter. Based on the established vulnerability of hepSTAT3-/- mice to secondary lung infections, we believe that intact liver function is critical for maintaining the immunological responsiveness of the lungs.


Subject(s)
Acute-Phase Reaction/immunology , Coinfection/immunology , Liver/metabolism , Lung/pathology , STAT3 Transcription Factor/metabolism , Airway Remodeling , Animals , Cells, Cultured , Endotoxemia , Inflammation , Lipopolysaccharides/metabolism , Liver/pathology , Mice , Mice, Knockout , Proteomics , STAT3 Transcription Factor/genetics , Transcriptome
7.
Infect Immun ; 89(4)2021 03 17.
Article in English | MEDLINE | ID: mdl-33526570

ABSTRACT

Pneumonia is a major public health concern, causing significant morbidity and mortality annually despite the broad use of antimicrobial agents. Underlying many of the severe sequelae of acute lung infections is dysfunction of the immune response, which remains incompletely understood yet is an attractive target of adjunct therapy in pneumonia. Here, we investigate the role of oncostatin M (OSM), a pleiotropic cytokine of the interleukin-6 (IL-6) family, and how its signaling modulates multiple innate immune pathways during pneumonia. Previously, we showed that OSM is necessary for neutrophil recruitment to the lungs during pneumonia by stimulating STAT3-driven CXCL5 expression. In this study, transcriptional profiling of whole-lung pneumonia with OSM neutralization revealed 241 differentially expressed genes following only 6 h of infection. Many downregulated genes are associated with STAT1, STAT3, and interferon signaling, suggesting these pathways are induced by OSM early in pneumonia. Interestingly, STAT1 and STAT3 activation was subsequently upregulated with OSM neutralization by 24 h, suggesting that OSM interruption dysregulates these central signaling pathways. When we investigated the source of OSM in pneumonia, neutrophils and, to a lesser extent, macrophages appear to be primary sources, suggesting a positive feedback loop of OSM production by neutrophils. From these studies, we conclude that OSM produced by recruited neutrophils tunes early innate immune signaling pathways, improving pneumonia outcomes.


Subject(s)
Neutrophils/immunology , Neutrophils/metabolism , Oncostatin M/metabolism , Pneumonia/etiology , Pneumonia/metabolism , Signal Transduction , Animals , Biomarkers , Cell Survival/genetics , Cell Survival/immunology , Gene Expression Regulation , Host-Pathogen Interactions/immunology , Immunity, Innate , Lung/immunology , Lung/metabolism , Lung/pathology , Macrophages/immunology , Macrophages/metabolism , Mice , Pneumonia/pathology
8.
JCI Insight ; 5(4)2020 02 27.
Article in English | MEDLINE | ID: mdl-31990682

ABSTRACT

Community-acquired pneumonia is a widespread disease with significant morbidity and mortality. Alveolar macrophages are tissue-resident lung cells that play a crucial role in innate immunity against bacteria that cause pneumonia. We hypothesized that alveolar macrophages display adaptive characteristics after resolution of bacterial pneumonia. We studied mice 1 to 6 months after self-limiting lung infections with Streptococcus pneumoniae, the most common cause of bacterial pneumonia. Alveolar macrophages, but not other myeloid cells, recovered from the lung showed long-term modifications of their surface marker phenotype. The remodeling of alveolar macrophages was (a) long-lasting (still observed 6 months after infection), (b) regionally localized (observed only in the affected lobe after lobar pneumonia), and (c) associated with macrophage-dependent enhanced protection against another pneumococcal serotype. Metabolomic and transcriptomic profiling revealed that alveolar macrophages of mice that recovered from pneumonia had new baseline activities and altered responses to infection that better resembled those of adult humans. The enhanced lung protection after mild and self-limiting bacterial respiratory infections includes a profound remodeling of the alveolar macrophage pool that is long-lasting; compartmentalized; and manifest across surface receptors, metabolites, and both resting and stimulated transcriptomes.


Subject(s)
Macrophages, Alveolar/immunology , Pneumonia, Pneumococcal/immunology , Animals , Cell Differentiation , Disease Models, Animal , Immunity, Innate , Lung/immunology , Mice , Myeloid Cells/cytology , Myeloid Cells/immunology
9.
PLoS One ; 14(8): e0221029, 2019.
Article in English | MEDLINE | ID: mdl-31415618

ABSTRACT

Interleukin-11 (IL-11) is an interleukin-6 (IL-6) family cytokine shown to play a protective role in acute inflammatory settings including systemic infection. In this study we addressed the role of IL-11 in acute bacterial pneumonia using a mouse model of E. coli pneumonia. Compared with other related cytokines, IL-11 protein was maintained at high levels in the lung at baseline, with only mild alterations in whole lung and BALF levels during acute infection. The primary source of IL-11 in the lung was the epithelium, but steady state production was not dependent on the inflammatory transcription factor nuclear factor kappa B in cells of either myeloid or epithelial lineage. Blockade of IL-11 with neutralizing antibodies resulted in a mild but significant decrease in neutrophil recruitment and increase in pulmonary edema during pneumonia, without detectable alterations in bacterial clearance. Exogenous IL-11 administration, however, had no effect at baseline or during infection. Overall, we conclude that maintenance of lung IL-11 concentrations may influence acute pulmonary inflammation during infection, albeit modestly.


Subject(s)
Interleukin-11/immunology , Neutrophil Infiltration/immunology , Neutrophils/immunology , Pneumonia, Bacterial/immunology , Pulmonary Edema/immunology , Acute Disease , Animals , Antibodies, Neutralizing/pharmacology , Interleukin-11/antagonists & inhibitors , Interleukin-11/genetics , Mice , Mice, Knockout , Neutrophil Infiltration/drug effects , Neutrophils/pathology , Pneumonia, Bacterial/drug therapy , Pneumonia, Bacterial/genetics , Pneumonia, Bacterial/pathology , Pulmonary Edema/drug therapy , Pulmonary Edema/genetics , Pulmonary Edema/pathology
10.
Infect Immun ; 87(8)2019 08.
Article in English | MEDLINE | ID: mdl-31160364

ABSTRACT

Pneumonia and sepsis are distinct but integrally linked public health concerns. The hepatic acute-phase response (APR), which is largely dependent on transcription factors NF-κB RelA and STAT3, is a hallmark of these pathologies and other injurious conditions. Inactivation of the APR can promote liver injury, a frequently observed organ dysfunction during sepsis. However, whether or how the acute-phase changes promote liver tissue resilience during infections is unclear. To determine the hepatoprotective role of the hepatic APR, we utilized mice bearing hepatocyte-specific deletions of either RelA or STAT3. Mice were challenged intratracheally (i.t.), intravenously (i.v.), or intraperitoneally (i.p.) with Escherichia coli, Klebsiella pneumoniae, Streptococcus pneumoniae, lipopolysaccharide (LPS), or alpha-galactosylceramide (αGalCer) to induce pneumonia, sepsis, or NKT cell activation. Liver injury was observed in RelA-null (hepRelAΔ/Δ) mice but not STAT3-null (hepSTAT3Δ/Δ) mice during pneumonia. The absence of RelA resulted in hepatotoxicity across several models of pneumonia, sepsis, and NKT cell activation. Injury was associated with increased levels of activated caspase-3 and -8 and substantial alteration of the hepatic transcriptome. Hepatotoxicity in the absence of RelA could be reversed by neutralization of tumor necrosis factor alpha (TNF-α). These results indicate the requirement of RelA-dependent inducible hepatoprotection during pneumonia and sepsis. Further, the results demonstrate that RelA-dependent gene programs are critical for maintaining liver homeostasis against TNF-α-driven immunotoxicity.


Subject(s)
Liver/pathology , Pneumonia/pathology , Sepsis/pathology , Transcription Factor RelA/physiology , Acute-Phase Reaction , Animals , Apoptosis , Chemokine CCL2/physiology , Kupffer Cells/physiology , Mice , Mice, Inbred C57BL , Natural Killer T-Cells/immunology , STAT3 Transcription Factor/physiology , Tumor Necrosis Factor-alpha/physiology
11.
Am J Physiol Lung Cell Mol Physiol ; 313(3): L548-L558, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28522567

ABSTRACT

In bacterial pneumonia, lung damage resulting from epithelial cell injury is a major contributor to the severity of disease and, in some cases, can lead to long-term sequelae, especially in the setting of severe lung injury or acute respiratory distress syndrome. Leukemia inhibitory factor (LIF), a member of the IL-6 cytokine family, is a critical determinant of lung tissue protection during pneumonia, but the cellular sources of LIF and the signaling pathways leading to its production in the infected lung are not known. Here, we demonstrate that lung epithelium, specifically alveolar type II cells, is the predominant site of LIF transcript induction in pneumonic mouse lungs. Epithelial cell cultures were induced to express LIF by bacteria and by sterile bronchoalveolar lavage fluid from pneumonic mice. Reciprocal bone marrow chimera studies demonstrated that LIF deficiency in the nonhematopoietic compartment, but not LIF deficiency in hematopoietic cells, eliminated LIF induction during pneumonia. Although NF-κB RelA (p65) is essential for the expression of many cytokines during pneumonia, its targeted mutation in the lung epithelium was inconsequential for pneumonia-driven LIF induction. However, maximal expression of this epithelial-derived cytokine was dependent on NF-κB RelA in myeloid cells. Overall, our data suggest a signaling axis whereby activation of NF-κB RelA in myeloid cells promotes epithelial LIF induction during lung infections, representing a means through which these two cell types collaborate to improve tissue resilience during pneumonia.


Subject(s)
Epithelial Cells/metabolism , Leukemia Inhibitory Factor/biosynthesis , Myeloid Cells/metabolism , Pneumonia, Bacterial/metabolism , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/pathology , Animals , Bronchoalveolar Lavage Fluid , Cell Line , Escherichia coli/physiology , Female , Hematopoiesis , Leukemia Inhibitory Factor/genetics , Lung/metabolism , Lung/pathology , Macrophages, Alveolar/metabolism , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Pneumonia, Bacterial/microbiology , Pneumonia, Bacterial/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription Factor RelA/metabolism
12.
Infect Immun ; 83(10): 4015-27, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26216424

ABSTRACT

Pneumonia and infection-induced sepsis are worldwide public health concerns. Both pathologies elicit systemic inflammation and induce a robust acute-phase response (APR). Although APR activation is well regarded as a hallmark of infection, the direct contributions of liver activation to pulmonary defense during sepsis remain unclear. By targeting STAT3-dependent acute-phase changes in the liver, we evaluated the role of liver STAT3 activity in promoting host defense in the context of sepsis and pneumonia. We employed a two-hit endotoxemia/pneumonia model, whereby administration of 18 h of intraperitoneal lipopolysaccharide (LPS; 5 mg/kg of body weight) was followed by intratracheal Escherichia coli (10(6) CFU) in wild-type mice or those lacking hepatocyte STAT3 (hepSTAT3(-/-)). Pneumonia alone (without endotoxemia) was effectively controlled in the absence of liver STAT3. Following endotoxemia and pneumonia, however, hepSTAT3(-/-) mice, with significantly reduced levels of circulating and airspace acute-phase proteins, exhibited significantly elevated lung and blood bacterial burdens and mortality. These data suggested that STAT3-dependent liver responses are necessary to promote host defense. While neither recruited airspace neutrophils nor lung injury was altered in endotoxemic hepSTAT3(-/-) mice, alveolar macrophage reactive oxygen species generation was significantly decreased. Additionally, bronchoalveolar lavage fluid from this group of hepSTAT3(-/-) mice allowed greater bacterial growth ex vivo. These results suggest that hepatic STAT3 activation promotes both cellular and humoral lung defenses. Taken together, induction of liver STAT3-dependent gene expression programs is essential to countering the deleterious consequences of sepsis on pneumonia susceptibility.


Subject(s)
Endotoxemia/immunology , Escherichia coli Infections/immunology , Liver/immunology , Lung/immunology , STAT3 Transcription Factor/immunology , Acute-Phase Reaction , Animals , Endotoxemia/genetics , Endotoxemia/microbiology , Escherichia coli/physiology , Escherichia coli Infections/genetics , Escherichia coli Infections/microbiology , Humans , Male , Mice , Pneumonia/genetics , Pneumonia/immunology , Pneumonia/microbiology , STAT3 Transcription Factor/genetics
13.
Am J Respir Cell Mol Biol ; 53(4): 479-88, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25692402

ABSTRACT

Acute bacterial pneumonia is a significant public health concern worldwide. Understanding the signals coordinating lung innate immunity may foster the development of therapeutics that limit tissue damage and promote host defense. We have previously shown that lung messenger RNA expression of the IL-6 family cytokine oncostatin-M (OSM) is significantly elevated in response to bacterial stimuli. However, its physiological significance during pneumonia is unknown. Here we demonstrate that OSM is rapidly increased in the airspaces of mice after pulmonary infection with Escherichia coli. Neutralization of OSM caused a substantial decrease in airspace neutrophils and macrophages. OSM blockade also caused a marked reduction in lung chemokine (C-X-C motif) ligand (CXCL) 5 expression, whereas other closely related neutrophil chemokines, CXCL1 and CXCL2, were unaffected. Intratracheal administration of recombinant OSM was sufficient to recapitulate the effect on CXCL5 induction, associated with robust activation of the signal transducer and activator of transcription 3 (STAT3) transcription factor. Cell sorting revealed that OSM effects were specific to lung epithelial cells, including a positive feedback loop in which OSM may facilitate expression of its own receptor. Finally, in vitro studies demonstrated that STAT3 was required for maximal OSM-induced CXCL5 expression. These studies demonstrate a novel role for OSM during pneumonia as an important signal to epithelial cells for chemokine induction mediating neutrophil recruitment.


Subject(s)
Chemokine CXCL5/metabolism , Escherichia coli Infections/metabolism , Oncostatin M/physiology , Pneumonia, Bacterial/metabolism , STAT3 Transcription Factor/metabolism , Animals , Cell Line, Tumor , Chemokine CXCL5/genetics , Escherichia coli Infections/immunology , Gene Expression , Humans , Mice, Inbred C57BL , Neutrophil Infiltration , Pneumonia, Bacterial/immunology , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism
14.
Am J Respir Cell Mol Biol ; 53(3): 378-90, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25607543

ABSTRACT

The hepatic acute-phase response (APR), stimulated by injury or inflammation, is characterized by significant changes in circulating acute-phase protein (APP) concentrations. Although individual functions of liver-derived APPs are known, the net consequence of APP changes is unclear. Pneumonia, which induces the APR, causes an inflammatory response within the airspaces that is coordinated largely by alveolar macrophages and is typified by cytokine production, leukocyte recruitment, and plasma extravasation, the latter of which may enable delivery of hepatocyte-derived APPs to the infection site. To determine the functional significance of the hepatic APR during pneumonia, we challenged APR-null mice lacking hepatocyte signal transducer and activator of transcription 3 (STAT3) and v-rel avian reticuloendotheliosis viral oncogene homolog A (RelA) with Escherichia coli in the airspaces. APR-null mice displayed ablated APP induction, significantly increased mortality, liver injury and apoptosis, and a trend toward increased bacterial burdens. TNF-α neutralization reversed hepatotoxicity, but not mortality, suggesting that APR-dependent survival is not solely due to hepatoprotection. After a milder (nonlethal) E. coli infection, hepatocyte-specific mutations decreased APP concentrations and pulmonary inflammation in bronchoalveolar lavage fluid. Cytokine expression in airspace macrophages, but not other airspace or circulating cells, was significantly dependent on APP extravasation into the alveoli. These data identify a novel signaling axis whereby the liver response enhances macrophage activation and pulmonary inflammation during pneumonia. Although hepatic acute-phase changes directly curb injury induced by TNF-α in the liver itself, APPs downstream of these same signals promote survival in association with innate immunity in the lungs, thus demonstrating a critical role for the lung-liver axis during pneumonia.


Subject(s)
Escherichia coli Infections/immunology , Liver/metabolism , Lung/metabolism , Pneumonia/immunology , Acute-Phase Proteins/metabolism , Animals , Cytokines/metabolism , Escherichia coli/immunology , Escherichia coli Infections/microbiology , Immunity, Innate , Liver/immunology , Lung/immunology , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Mice , Mice, Transgenic , Pneumonia/microbiology
15.
Microbiology (Reading) ; 154(Pt 8): 2265-2274, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18667559

ABSTRACT

The accessory gene regulator (agr) of Staphylococcus aureus is a global regulator of the staphylococcal virulon, which includes secreted virulence factors and surface proteins. The agr locus is important for virulence in a variety of animal models of infection, and has been assumed by inference to have a major role in human infection. Although most human clinical S. aureus isolates are agr(+), there have been several reports of agr-defective mutants isolated from infected patients. Since it is well known that the agr locus is genetically labile in vitro, we have addressed the question of whether the reported agr-defective mutants were involved in the infection or could have arisen during post-isolation handling. We obtained a series of new staphylococcal isolates from local clinical infections and handled these with special care to avoid post-isolation mutations. Among these isolates, we found a number of strains with non-haemolytic phenotypes owing to mutations in the agr locus, and others with mutations elsewhere. We have also obtained isolates in which the population was continuously heterogeneous with respect to agr functionality, with agr(+) and agr(-) variants having otherwise indistinguishable chromosomal backgrounds. This finding suggested that the agr(-) variants arose by mutation during the course of the infection. Our results indicate that while most clinical isolates are haemolytic and agr(+), non-haemolytic and agr(-) strains are found in S. aureus infections, and that agr(+) and agr(-) variants may have a cooperative interaction in certain types of infections.


Subject(s)
Bacterial Proteins/metabolism , Staphylococcal Infections/microbiology , Staphylococcus aureus/isolation & purification , Staphylococcus aureus/metabolism , Trans-Activators/metabolism , Amino Acid Sequence , Bacterial Proteins/genetics , Bacterial Typing Techniques , Base Sequence , Genotype , Hemolysin Proteins/genetics , Hemolysin Proteins/metabolism , Humans , Molecular Sequence Data , Mutation , Staphylococcus aureus/classification , Staphylococcus aureus/genetics , Trans-Activators/genetics , Transcription, Genetic
16.
J Bacteriol ; 187(16): 5585-94, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16077103

ABSTRACT

Agr is a global regulatory system in the staphylococci, operating by a classical two-component signaling module and controlling the expression of most of the genes encoding extracellular virulence factors. As it is autoinduced by a peptide, encoded within the locus, that is the ligand for the signal receptor, it is a sensor of population density or a quorum sensor and is the only known quorum-sensing system in the genus. agr is conserved throughout the staphylococci but has diverged along lines that appear to parallel speciation and subspeciation within the genus. This divergence has given rise to a novel type of interstrain and interspecies cross-inhibition that represents a fundamental aspect of the organism's biology and may be a predominant feature of the evolutionary forces that have driven it. We present evidence, using a newly developed, luciferase-based agr typing scheme, that the evolutionary divergence of the agr system was an early event in the evolution of the staphylococci and long preceded the development of the nucleotide polymorphisms presently used for genotyping. These polymorphisms developed, for the most part, within different agr groups; mobile genetic elements appear also to have diffused recently and, with a few notable exceptions, have come to reside largely indiscriminately within the several agr groups.


Subject(s)
Bacterial Proteins/genetics , Evolution, Molecular , Gene Expression Regulation, Bacterial , Staphylococcus aureus/genetics , Trans-Activators/genetics , Genetic Variation , Genotype , Hemolysin Proteins/metabolism , Hemolysis , Luciferases/genetics , Phenotype , Promoter Regions, Genetic/physiology , Staphylococcus aureus/metabolism , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...