Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-38656811

ABSTRACT

Pseudomonas aeruginosa causes chronic lung infection in cystic fibrosis (CF), resulting in structural lung damage and progressive pulmonary decline. P. aeruginosa in the CF lung undergoes numerous changes, adapting to host-specific airway pressures while establishing chronic infection. P. aeruginosa undergoes lipid A structural modification during CF chronic infection, not seen in any other disease state. Lipid A, the membrane anchor of lipopolysaccharide (i.e., endotoxin), comprises the majority of the outer membrane of Gram-negative bacteria and is a potent toll-like receptor (TLR)4 agonist. The structure of P. aeruginosa lipid A is intimately linked with its recognition by TLR4, and subsequent immune response. Prior work has identified P. aeruginosa strains with altered lipid A structures that arise during chronic CF lung infection; however, the impact of P. aeruginosa lipid A structure on airway disease has not been investigated. Here, we show that P. aeruginosa lipid A lacks PagL-mediated deacylation during human airway infection using a direct-from-sample mass spectrometry approach on human bronchoalveolar lavage fluid. This structure triggers increased pro-inflammatory cytokine production by primary human macrophages. Furthermore, alterations in lipid A 2-hydroxylation impact cytokine response in a site-specific manner, independent of CFTR function. Interestingly, there is a CF-specific reduction in IL-8 secretion within the epithelial-cell compartment that only occurs in CF bronchial epithelial cells when infected with CF-adapted P. aeruginosa that lack PagL-mediated lipid A deacylation. Taken together, we show that P. aeruginosa alters its lipid A structure during acute lung infection and that this lipid A structure induces stronger signaling through TLR4.

2.
J Innate Immun ; 16(1): 143-158, 2024.
Article in English | MEDLINE | ID: mdl-38310854

ABSTRACT

BACKGROUND: Upon infection, mucosal tissues activate a brisk inflammatory response to clear the pathogen, i.e., resistance to disease. Resistance to disease is orchestrated by tissue-resident macrophages, which undergo profound metabolic reprogramming after sensing the pathogen. These metabolically activated macrophages release many inflammatory factors, which promote their bactericidal function. However, in immunocompetent individuals, pathogens like Pseudomonas aeruginosa, Staphylococcus aureus, and Salmonella evade this type of immunity, generating communities that thrive for the long term. SUMMARY: These organisms develop features that render them less susceptible to eradication, such as biofilms and increased tolerance to antibiotics. Furthermore, after antibiotic therapy withdrawal, "persister" cells rapidly upsurge, triggering inflammatory relapses that worsen host health. How these pathogens persisted in inflamed tissues replete with activated macrophages remains poorly understood. KEY MESSAGES: In this review, we discuss recent findings indicating that the ability of P. aeruginosa, S. aureus, and Salmonella to evolve biofilms and antibiotic tolerance is promoted by the similar metabolic routes that regulate macrophage metabolic reprogramming.


Subject(s)
Anti-Bacterial Agents , Biofilms , Macrophages , Biofilms/drug effects , Humans , Animals , Macrophages/immunology , Macrophages/microbiology , Anti-Bacterial Agents/pharmacology , Bacterial Infections/immunology , Pseudomonas aeruginosa/immunology , Pseudomonas aeruginosa/physiology , Staphylococcus aureus/immunology , Staphylococcus aureus/physiology , Drug Resistance, Bacterial , Immune Evasion
3.
Cell Metab ; 35(10): 1767-1781.e6, 2023 10 03.
Article in English | MEDLINE | ID: mdl-37793346

ABSTRACT

Pseudomonas aeruginosa is a common cause of pulmonary infection. As a Gram-negative pathogen, it can initiate a brisk and highly destructive inflammatory response; however, most hosts become tolerant to the bacterial burden, developing chronic infection. Using a murine model of pneumonia, we demonstrate that this shift from inflammation to disease tolerance is promoted by ketogenesis. In response to pulmonary infection, ketone bodies are generated in the liver and circulate to the lungs where they impose selection for P. aeruginosa strains unable to display surface lipopolysaccharide (LPS). Such keto-adapted LPS strains fail to activate glycolysis and tissue-damaging cytokines and, instead, facilitate mitochondrial catabolism of fats and oxidative phosphorylation (OXPHOS), which maintains airway homeostasis. Within the lung, P. aeruginosa exploits the host immunometabolite itaconate to further stimulate ketogenesis. This environment enables host-P. aeruginosa coexistence, supporting both pathoadaptive changes in the bacteria and the maintenance of respiratory integrity via OXPHOS.


Subject(s)
Lipopolysaccharides , Pseudomonas aeruginosa , Mice , Animals , Lung , Inflammation , Ketone Bodies
4.
Cell Rep ; 42(2): 112064, 2023 02 28.
Article in English | MEDLINE | ID: mdl-36724077

ABSTRACT

Neutrophils are critical in the host defense against Staphylococcus aureus, a major human pathogen. However, even in the setting of a robust neutrophil response, S. aureus can evade immune clearance. Here, we demonstrate that S. aureus impairs neutrophil function by triggering the production of the anti-inflammatory metabolite itaconate. The enzyme that synthesizes itaconate, Irg1, is selectively expressed in neutrophils during S. aureus pneumonia. Itaconate inhibits neutrophil glycolysis and oxidative burst, which impairs survival and bacterial killing. In a murine pneumonia model, neutrophil Irg1 expression protects the lung from excessive inflammation but compromises bacterial clearance. S. aureus is thus able to evade the innate immune response by targeting neutrophil metabolism and inducing the production of the anti-inflammatory metabolite itaconate.


Subject(s)
Staphylococcal Infections , Staphylococcus aureus , Humans , Animals , Mice , Neutrophils/metabolism , Respiratory Burst , Staphylococcal Infections/microbiology
5.
Clin Chest Med ; 43(4): 647-665, 2022 12.
Article in English | MEDLINE | ID: mdl-36344072

ABSTRACT

Highly effective cystic fibrosis (CF) transmembrane conductance regulator (CFTR) modulator therapy (HEMT) corrects the underlying molecular defect causing CF disease. HEMT decreases symptom burden and improves clinical metrics and quality of life for most people with CF (PwCF) and eligible cftr mutations. Improvements in measures of pulmonary health suggest that restoration of function of defective CFTR anion channels by HEMT not only enhances airway mucociliary clearance, but also reduces chronic pulmonary infection and inflammation. This article reviews the evidence for how HEMT influences the dynamic and interdependent processes of infection and inflammation in the CF airway, and what questions remain unanswered.


Subject(s)
Cystic Fibrosis , Quinolones , Humans , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/therapeutic use , Cystic Fibrosis/drug therapy , Aminophenols/therapeutic use , Quality of Life , Quinolones/therapeutic use , Mutation , Inflammation/drug therapy
6.
Pathogens ; 10(8)2021 Jul 29.
Article in English | MEDLINE | ID: mdl-34451421

ABSTRACT

Prevailing dogma indicates that the lung of cystic fibrosis (CF) individuals is infected by multiple pathogens due to the abundant accumulation of mucus, which traps most of inhaled organisms. However, this hypothesis does not explain how specific opportunists, like Pseudomonas aeruginosa, are selected in the CF lung to cause chronic disease. This strongly suggests that other factors than mucus are accrued in the human airway and might predispose to bacterial disease, especially by P. aeruginosa. In this review we discuss the role of macrophage metabolites, like succinate and itaconate, in P. aeruginosa pneumonia. We analyze how dysfunction of the CF transmembrane conductance regulator (CFTR) favors release of these metabolites into the infected airway, and how P. aeruginosa exploits these elements to induce transcriptomic and metabolic changes that increase its capacity to cause intractable disease. We describe the host and pathogen pathways associated with succinate and itaconate catabolism, mechanisms of bacterial adaptation to these determinants, and suggest how both experimental settings and future therapies should consider macrophage metabolites abundance to better study P. aeruginosa pathogenesis.

7.
Microb Cell ; 8(5): 106-107, 2021 Apr 19.
Article in English | MEDLINE | ID: mdl-33981762

ABSTRACT

Staphylococcus aureus is a prominent pathogen that can cause intractable lung infections in humans. S. aureus persists in the airway despite inflammation and immune cell recruitment by adapting to host-derived antimicrobial factors. A key component of the immune response to infection are host metabolites that regulate inflammation and bacterial survival. In our recent paper (Tomlinson et al., Nat Commun, doi: 10.1038/s41467-021-21718-y), we demonstrated that S. aureus induces the production of the immunoregulatory metabolite itaconate in airway immune cells by stimulating mitochondrial oxidant stress. Itaconate in turn inhibited S. aureus glycolysis and growth, and promoted carbon flux through bacterial metabolic pathways that support biofilm production. These itaconate-induced metabolic changes were recapitulated in a longitudinal series of clinical isolates from a patient with chronic staphylococcal lung infections, demonstrating a role for host immunometabolism in driving bacterial persistence during long-term staphylococcal lung infections.

8.
Nat Commun ; 12(1): 1399, 2021 03 03.
Article in English | MEDLINE | ID: mdl-33658521

ABSTRACT

Staphylococcus aureus is a prominent human pathogen that readily adapts to host immune defenses. Here, we show that, in contrast to Gram-negative pathogens, S. aureus induces a distinct airway immunometabolic response dominated by the release of the electrophilic metabolite, itaconate. The itaconate synthetic enzyme, IRG1, is activated by host mitochondrial stress, which is induced by staphylococcal glycolysis. Itaconate inhibits S. aureus glycolysis and selects for strains that re-direct carbon flux to fuel extracellular polysaccharide (EPS) synthesis and biofilm formation. Itaconate-adapted strains, as illustrated by S. aureus isolates from chronic airway infection, exhibit decreased glycolytic activity, high EPS production, and proficient biofilm formation even before itaconate stimulation. S. aureus thus adapts to the itaconate-dominated immunometabolic response by producing biofilms, which are associated with chronic infection of the human airway.


Subject(s)
Host-Pathogen Interactions/physiology , Staphylococcal Infections/immunology , Staphylococcus aureus/physiology , Staphylococcus aureus/pathogenicity , Succinates/metabolism , Adult , Animals , Biofilms/growth & development , Bronchoalveolar Lavage Fluid , Carbohydrate Metabolism , Cystic Fibrosis/microbiology , Gene Expression Regulation, Bacterial , Glycolysis/drug effects , Glycolysis/physiology , Host-Pathogen Interactions/immunology , Humans , Hydro-Lyases/metabolism , Mice, Inbred C57BL , Pseudomonas Infections/immunology , Pseudomonas Infections/metabolism , Reactive Oxygen Species/metabolism , Sputum/microbiology , Staphylococcal Infections/metabolism , Staphylococcus aureus/drug effects , Staphylococcus aureus/isolation & purification , Stress, Physiological , Succinates/pharmacology , Succinic Acid/metabolism , Young Adult
9.
Respir Res ; 21(1): 326, 2020 Dec 10.
Article in English | MEDLINE | ID: mdl-33302964

ABSTRACT

Pulmonary infections are associated with a brisk inflammatory reaction to bacterial surface components. Lipopolysaccharides (LPS) trigger macrophage activation and release of mitochondrial metabolites that control the intensity of the immune response. Whereas succinate induces oxidative stress (ROS), HIF1α stabilization, glycolysis and IL-1ß release, itaconate suppresses inflammation by inhibiting succinate oxidation, glycolytic flux and promoting anti-oxidant Nrf2-HO-1 functions. P. aeruginosa is a major pathogen associated with acute and chronic lung infection. Although both secreted toxins, LPS and proteases are key factors to establish acute P. aeruginosa pneumonia, lack of these components in chronic P. aeruginosa isolates suggest these organisms exploit other mechanisms to adapt and persist in the lung. Upon inhalation, P. aeruginosa strains trigger airway macrophage reprograming and bacterial variants obtained from acutely and chronically infected subjects exhibit metabolic adaptation consistent with succinate and itaconate assimilation; namely, high expression of extracellular polysaccharides (EPS), reduced lptD-LPS function, increased glyoxylate shunt (GS) activity and substantial biofilm production. In this review we discuss recent findings illustrating how P. aeruginosa induces and adapts to macrophage metabolites in the human lung, and that catabolism of succinate and itaconate contribute to their formidable abilities to tolerate oxidative stress, phagocytosis and immune clearance.


Subject(s)
Energy Metabolism , Lung/microbiology , Macrophage Activation , Macrophages, Alveolar/microbiology , Pneumonia, Bacterial/microbiology , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/pathogenicity , Animals , Biofilms/growth & development , Host-Pathogen Interactions , Humans , Inflammation Mediators/metabolism , Lung/immunology , Lung/metabolism , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Oxidative Stress , Pneumonia, Bacterial/immunology , Pneumonia, Bacterial/metabolism , Pseudomonas Infections/immunology , Pseudomonas Infections/metabolism , Pseudomonas aeruginosa/growth & development , Pseudomonas aeruginosa/immunology , Reactive Oxygen Species/metabolism
10.
Cell Metab ; 31(6): 1091-1106.e6, 2020 06 02.
Article in English | MEDLINE | ID: mdl-32428444

ABSTRACT

The bacterium Pseudomonas aeruginosa is especially pathogenic, often being associated with intractable pneumonia and high mortality. How P. aeruginosa avoids immune clearance and persists in the inflamed human airway remains poorly understood. In this study, we show that P. aeruginosa can exploit the host immune response to maintain infection. Notably, unlike other opportunistic bacteria, we found that P. aeruginosa alters its metabolic and immunostimulatory properties in response to itaconate, an abundant host-derived immunometabolite in the infected lung. Itaconate induces bacterial membrane stress, resulting in downregulation of lipopolysaccharides (LPS) and upregulation of extracellular polysaccharides (EPS). These itaconate-adapted P. aeruginosa accumulate lptD mutations, which favor itaconate assimilation and biofilm formation. EPS, in turn, induces itaconate production by myeloid cells, both in the airway and systemically, skewing the host immune response to one permissive of chronic infection. Thus, the metabolic versatility of P. aeruginosa needs to be taken into account when designing therapies.


Subject(s)
Biofilms , Pseudomonas aeruginosa/metabolism , Succinates/metabolism , Animals , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout
11.
Front Immunol ; 11: 385, 2020.
Article in English | MEDLINE | ID: mdl-32231665

ABSTRACT

A limited number of pulmonary pathogens are able to evade normal mucosal defenses to establish acute infection and then adapt to cause chronic pneumonias. Pathogens, such as Pseudomonas aeruginosa or Staphylococcus aureus, are typically associated with infection in patients with underlying pulmonary disease or damage, such as cystic fibrosis (CF) or chronic obstructive pulmonary disease (COPD). To establish infection, bacteria express a well-defined set of so-called virulence factors that facilitate colonization and activate an immune response, gene products that have been identified in murine models. Less well-understood are the adaptive changes that occur over time in vivo, enabling the organisms to evade innate and adaptive immune clearance mechanisms. These colonizers proliferate, generating a population sufficient to provide selection for mutants, such as small colony variants and mucoid variants, that are optimized for long term infection. Such host-adapted strains have evolved in response to selective pressure such as antibiotics and the recruitment of phagocytes at sites of infection and their release of signaling metabolites (e.g., succinate). These metabolites can potentially function as substrates for bacterial growth and but also generate oxidant stress. Whole genome sequencing and quantified expression of selected genes have helped to explain how P. aeruginosa and S. aureus adapt to the presence of these metabolites over the course of in vivo infection. The serial isolation of clonally related strains from patients with cystic fibrosis has provided the opportunity to identify bacterial metabolic pathways that are altered under this immune pressure, such as the anti-oxidant glyoxylate and pentose phosphate pathways, routes contributing to the generation of biofilms. These metabolic pathways and biofilm itself enable the organisms to dissipate oxidant stress, while providing protection from phagocytosis. Stimulation of host immune signaling metabolites by these pathogens drives bacterial adaptation and promotes their persistence in the airways. The inherent metabolic flexibility of P. aeruginosa and S. aureus is a major factor in their success as pulmonary pathogens.


Subject(s)
Host-Pathogen Interactions/immunology , Pseudomonas aeruginosa/physiology , Respiratory Tract Infections/immunology , Respiratory Tract Infections/microbiology , Staphylococcus aureus/physiology , Adaptation, Physiological/immunology , Animals , Biofilms , Chronic Disease , Humans , Pseudomonas Infections/immunology , Pseudomonas aeruginosa/immunology , Pseudomonas aeruginosa/pathogenicity , Staphylococcal Infections/immunology , Staphylococcus aureus/immunology , Staphylococcus aureus/pathogenicity
12.
Nat Microbiol ; 5(1): 141-153, 2020 01.
Article in English | MEDLINE | ID: mdl-31686028

ABSTRACT

Staphylococcus aureus small colony variants (SCVs) are frequently associated with chronic infection, yet they lack expression of many virulence determinants associated with the pathogenicity of wild-type strains. We found that both wild-type S. aureus and a ΔhemB SCV prototype potently activate glycolysis in host cells. Glycolysis and the generation of mitochondrial reactive oxygen species were sufficient to induce necroptosis, a caspase-independent mechanism of host cell death that failed to eradicate S. aureus and instead promoted ΔhemB SCV pathogenicity. To support ongoing glycolytic activity, the ΔhemB SCV induced over a 100-fold increase in the expression of fumC, which encodes an enzyme that catalyses the degradatin of fumarate, an inhibitor of glycolysis. Consistent with fumC-dependent depletion of local fumarate, the ΔhemB SCV failed to elicit trained immunity and protection from a secondary infectious challenge in the skin. The reliance of the S. aureus SCV population on glycolysis accounts for much of its role in the pathogenesis of S. aureus skin infection.


Subject(s)
Immunomodulation , Staphylococcal Skin Infections/metabolism , Staphylococcal Skin Infections/pathology , Staphylococcus aureus/pathogenicity , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cells, Cultured , Fumarates/metabolism , Gene Expression Regulation, Bacterial , Glycolysis , Humans , Immune Evasion , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Necroptosis/genetics , Reactive Oxygen Species/metabolism , Staphylococcal Skin Infections/immunology , Staphylococcal Skin Infections/microbiology , Staphylococcus aureus/genetics , Staphylococcus aureus/growth & development , Staphylococcus aureus/metabolism , THP-1 Cells
13.
Sci Transl Med ; 11(499)2019 07 03.
Article in English | MEDLINE | ID: mdl-31270271

ABSTRACT

Phosphatase and tensin homolog deleted on chromosome 10 (PTEN) is a tumor suppressor best known for regulating cell proliferation and metabolism. PTEN forms a complex with the cystic fibrosis (CF) transmembrane conductance regulator (CFTR) at the plasma membrane, and this complex is known to be functionally impaired in CF. Here, we demonstrated that the combined effect of PTEN and CFTR dysfunction stimulates mitochondrial activity, resulting in excessive release of succinate and reactive oxygen species. This environment promoted the colonization of the airway by Pseudomonas aeruginosa, bacteria that preferentially metabolize succinate, and stimulated an anti-inflammatory host response dominated by immune-responsive gene 1 (IRG1) and itaconate. The recruitment of myeloid cells induced by these strains was inefficient in clearing the infection and increased numbers of phagocytes accumulated under CFTR-PTEN axis dysfunction. This central metabolic defect in mitochondrial function due to impaired PTEN activity contributes to P. aeruginosa infection in CF.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Lung/microbiology , Mitochondria/metabolism , PTEN Phosphohydrolase/metabolism , Pseudomonas Infections/metabolism , Animals , Carboxy-Lyases/metabolism , Colony Count, Microbial , Cystic Fibrosis/pathology , HCT116 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunity , Interleukin-1beta/metabolism , Lung/immunology , Mice, Inbred C57BL , Middle Aged , Oxidants/metabolism , Oxidative Stress , PTEN Phosphohydrolase/deficiency , Pseudomonas aeruginosa/isolation & purification , Reactive Oxygen Species/metabolism , Succinates/metabolism
14.
Am J Respir Cell Mol Biol ; 61(2): 185-197, 2019 08.
Article in English | MEDLINE | ID: mdl-30742488

ABSTRACT

Methicillin-resistant Staphylococcus aureus (MRSA) is a versatile human pathogen that is associated with diverse types of infections ranging from benign colonization to sepsis. We postulated that MRSA must undergo specific genotypic and phenotypic changes to cause chronic pulmonary disease. We investigated how MRSA adapts to the human airway to establish chronic infection, as occurs during cystic fibrosis (CF). MRSA isolates from patients with CF that were collected over a 4-year period were analyzed by whole-genome sequencing, transcriptional analysis, and metabolic studies. Persistent MRSA infection was associated with staphylococcal metabolic adaptation, but not changes in immunogenicity. Adaptation was characterized by selective use of the tricarboxylic acid cycle cycle and generation of biofilm, a means of limiting oxidant stress. Increased transcription of specific metabolic genes was conserved in all host-adapted strains, most notably a 10,000-fold increase in fumC, which catalyzes the interconversion of fumarate and malate. Elevated fumarate levels promoted in vitro biofilm production in clinical isolates. Host-adapted strains preferred to assimilate glucose polymers and pyruvate, which can be metabolized to generate N-acetylglucosamine polymers that comprise biofilm. MRSA undergoes substantial metabolic adaptation to the human airway to cause chronic pulmonary infection, and selected metabolites may be useful therapeutically to inhibit infection.


Subject(s)
Cystic Fibrosis/microbiology , Lung Diseases/microbiology , Methicillin-Resistant Staphylococcus aureus/metabolism , Pneumonia, Staphylococcal/microbiology , Staphylococcal Infections/microbiology , Acetylglucosamine/metabolism , Adult , Animals , Biofilms , Bronchi/metabolism , Bronchoalveolar Lavage Fluid , Cystic Fibrosis/metabolism , Cytokines/metabolism , Female , Fumarates/metabolism , Gentamicins/pharmacology , Glucose/metabolism , Humans , Lung Diseases/metabolism , Malates/metabolism , Male , Mice , Mice, Inbred C57BL , Middle Aged , Phylogeny , Pneumonia, Staphylococcal/metabolism , Pyruvic Acid/metabolism , Staphylococcal Infections/metabolism , Transcription, Genetic , Tricarboxylic Acids/metabolism , Whole Genome Sequencing
15.
J Innate Immun ; 10(5-6): 442-454, 2018.
Article in English | MEDLINE | ID: mdl-29617698

ABSTRACT

Many different species of gram-negative bacteria are associated with infection in the lung, causing exacerbations of chronic obstructive pulmonary disease, cystic fibrosis (CF), and ventilator-associated pneumonias. These airway pathogens must adapt to common host clearance mechanisms that include killing by antimicrobial peptides, antibiotics, oxidative stress, and phagocytosis by leukocytes. Bacterial adaptation to the host is often evident phenotypically, with increased extracellular polysaccharide production characteristic of some biofilm-associated organisms. Given the relatively limited repertoire of bacterial strategies to elude airway defenses, it seems likely that organisms sharing the same ecological niche might also share common strategies to persistently infect the lung. In this review, we will highlight some of the major factors responsible for the adaptation of Pseudomonas aeruginosa to the lung, addressing how growth in biofilms enables persistent infection, relevant to, but not limited to, the pathogenesis of infection in CF. In contrast, we will discuss how carbapenem-resistant Klebsiella pneumoniae evade immune clearance, an organism often associated with ventilator-associated pneumonia and health-care-acquired pneumonias, but not a typical pathogen in CF.


Subject(s)
Cystic Fibrosis/immunology , Klebsiella Infections/immunology , Klebsiella pneumoniae/physiology , Lung/immunology , Pseudomonas Infections/immunology , Pseudomonas aeruginosa/physiology , Animals , Biofilms , Cystic Fibrosis/microbiology , Host-Pathogen Interactions , Humans , Immune Evasion , Immunity, Innate , Lung/microbiology
16.
Immunity ; 47(6): 1169-1181.e7, 2017 12 19.
Article in English | MEDLINE | ID: mdl-29246444

ABSTRACT

The tumor suppressor PTEN controls cell proliferation by regulating phosphatidylinositol-3-kinase (PI3K) activity, but the participation of PTEN in host defense against bacterial infection is less well understood. Anti-inflammatory PI3K-Akt signaling is suppressed in patients with cystic fibrosis (CF), a disease characterized by hyper-inflammatory responses to airway infection. We found that Ptenl-/- mice, which lack the NH2-amino terminal splice variant of PTEN, were unable to eradicate Pseudomonas aeruginosa from the airways and could not generate sufficient anti-inflammatory PI3K activity, similar to what is observed in CF. PTEN and the CF transmembrane conductance regulator (CFTR) interacted directly and this interaction was necessary to position PTEN at the membrane. CF patients under corrector-potentiator therapy, which enhances CFTR transport to the membrane, have increased PTEN amounts. These findings suggest that improved CFTR trafficking could enhance P. aeruginosa clearance from the CF airway by activating PTEN-mediated anti-bacterial responses and might represent a therapeutic strategy.


Subject(s)
Cell Membrane/immunology , Cystic Fibrosis Transmembrane Conductance Regulator/immunology , Cystic Fibrosis/immunology , PTEN Phosphohydrolase/immunology , Pseudomonas Infections/immunology , Aminophenols/pharmacology , Aminopyridines/pharmacology , Animals , Benzodioxoles/pharmacology , Cell Membrane/drug effects , Cystic Fibrosis/drug therapy , Cystic Fibrosis/genetics , Cystic Fibrosis/microbiology , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Gene Expression Regulation , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Molecular , Monocytes/drug effects , Monocytes/immunology , Monocytes/microbiology , PTEN Phosphohydrolase/deficiency , PTEN Phosphohydrolase/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/immunology , Protein Binding , Protein Conformation , Protein Transport , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/immunology , Pseudomonas Infections/genetics , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/immunology , Quinolones/pharmacology , Signal Transduction
17.
J Immunol ; 199(1): 212-223, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28566367

ABSTRACT

Human respiratory syncytial virus (hRSV) is the leading cause of severe lower respiratory tract infections in children. The development of novel prophylactic and therapeutic antiviral drugs against hRSV is imperative to control the burden of disease in the susceptible population. In this study, we examined the effects of inducing the activity of the host enzyme heme oxygenase-1 (HO-1) on hRSV replication and pathogenesis on lung inflammation induced by this virus. Our results show that after hRSV infection, HO-1 induction with metalloporphyrin cobalt protoporphyrin IX significantly reduces the loss of body weight due to hRSV-induced disease. Further, HO-1 induction also decreased viral replication and lung inflammation, as evidenced by a reduced neutrophil infiltration into the airways, with diminished cytokine and chemokine production and reduced T cell function. Concomitantly, upon cobalt protoporphyrin IX treatment, there is a significant upregulation in the production of IFN-α/ß mRNAs in the lungs. Furthermore, similar antiviral and protective effects occur by inducing the expression of human HO-1 in MHC class II+ cells in transgenic mice. Finally, in vitro data suggest that HO-1 induction can modulate the susceptibility of cells, especially the airway epithelial cells, to hRSV infection.


Subject(s)
Heme Oxygenase-1/metabolism , Lung/immunology , Respiratory Syncytial Virus Infections/physiopathology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus, Human/physiology , Animals , Cell Line , Cytokines/biosynthesis , Cytokines/immunology , DNA Replication , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Heme Oxygenase-1/genetics , Humans , Interferon-alpha/biosynthesis , Interferon-alpha/immunology , Interferon-beta/immunology , Lung/metabolism , Lung/pathology , Lung/virology , Mice , Protoporphyrins/administration & dosage , Protoporphyrins/pharmacology , Respiratory Syncytial Virus Infections/immunology , T-Lymphocytes/immunology , Virus Attachment , Virus Internalization , Virus Replication
18.
Immunology ; 149(1): 1-12, 2016 09.
Article in English | MEDLINE | ID: mdl-26938875

ABSTRACT

Haem-oxygenase-1 (HO-1) is an enzyme responsible for the degradation of haem that can suppress inflammation, through the production of carbon monoxide (CO). It has been shown in several experimental models that genetic and pharmacological induction of HO-1, as well as non-toxic administration of CO, can reduce inflammatory diseases, such as endotoxic shock, type 1 diabetes and graft rejection. Recently, it was shown that the HO-1/CO system can alter the function of antigen-presenting cells (APCs) and reduce T-cell priming, which can be beneficial during immune-driven inflammatory diseases. The molecular mechanisms by which the HO-1 and CO reduce both APC- and T-cell-driven immunity are just beginning to be elucidated. In this article we discuss recent findings related to the immune regulatory capacity of HO-1 and CO at the level of recognition of pathogen-associated molecular patterns and T-cell priming by APCs. Finally, we propose a possible regulatory role for HO-1 and CO over the recently described mitochondria-dependent immunity. These concepts could contribute to the design of new therapeutic tools for inflammation-based diseases.


Subject(s)
Antigen Presentation , Heme Oxygenase-1/metabolism , Immune System Diseases/drug therapy , Immune Tolerance , Inflammation/metabolism , T-Lymphocytes/immunology , Animals , Carbon Monoxide/metabolism , Drug Design , Humans , Immunomodulation , Lymphocyte Activation
19.
Immunology ; 147(1): 55-72, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26451966

ABSTRACT

Human respiratory syncytial virus (hRSV) is the leading cause of infant hospitalization related to respiratory disease. Infection with hRSV produces abundant infiltration of immune cells into the airways, which combined with an exacerbated pro-inflammatory immune response can lead to significant damage to the lungs. Human RSV re-infection is extremely frequent, suggesting that this virus may have evolved molecular mechanisms that interfere with host adaptive immunity. Infection with hRSV can be reduced by administering a humanized neutralizing antibody against the virus fusion protein in high-risk infants. Although neutralizing antibodies against hRSV effectively block the infection of airway epithelial cells, here we show that both, bone marrow-derived dendritic cells (DCs) and lung DCs undergo infection with IgG-coated virus (hRSV-IC), albeit abortive. Yet, this is enough to negatively modulate DC function. We observed that such a process is mediated by Fcγ receptors (FcγRs) expressed on the surface of DCs. Remarkably, we also observed that in the absence of hRSV-specific antibodies FcγRIII knockout mice displayed significantly less cellular infiltration in the lungs after hRSV infection, compared with wild-type mice, suggesting a potentially harmful, IgG-independent role for this receptor in hRSV disease. Our findings support the notion that FcγRs can contribute significantly to the modulation of DC function by hRSV and hRSV-IC. Further, we provide evidence for an involvement of FcγRIII in the development of hRSV pathogenesis.


Subject(s)
Dendritic Cells/metabolism , Lung/metabolism , Lymphocyte Activation , Receptors, IgG/metabolism , Respiratory Syncytial Virus Infections/metabolism , Respiratory Syncytial Virus, Human/pathogenicity , T-Lymphocytes/metabolism , Adaptive Immunity , Animals , Antibodies, Neutralizing/pharmacology , Antibodies, Viral/immunology , Antibodies, Viral/metabolism , Antiviral Agents/pharmacology , Cells, Cultured , Coculture Techniques , Cytokines/metabolism , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/virology , Disease Models, Animal , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Lung/drug effects , Lung/immunology , Lung/virology , Lymphocyte Activation/drug effects , Mice, Inbred C57BL , Mice, Knockout , Palivizumab/pharmacology , Receptors, IgG/deficiency , Receptors, IgG/genetics , Receptors, IgG/immunology , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Virus Infections/genetics , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus, Human/drug effects , Respiratory Syncytial Virus, Human/immunology , Signal Transduction , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/virology , Viral Load , Virus Replication
20.
Eur J Immunol ; 45(12): 3269-88, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26461179

ABSTRACT

Heme-oxygenase 1 (HO-1) prevents T cell-mediated inflammatory disease by producing carbon monoxide (CO) and impairing DC immunogenicity. However, the cellular mechanisms causing this inhibition are unknown. Here, we show that CO impairs mitochondrial function in DCs by reducing both the mitochondrial membrane potential and ATP production, and resembling the effect of a nonlethal dose of a classical mitochondria uncoupler carbonyl cyanide m-chlorophenyl hydrazone (CCCP). Moreover, both CO and CCCP reduced cargo transport, endosome-to-lysosome fusion, and antigen processing, dampening the production of peptide-MHC complexes on the surface of DCs. As a result, the inhibition of naive CD4(+) T-cell priming was observed. Furthermore, mitochondrial dysfunction in DCs also significantly reduced CD8(+) T cell-dependent type 1 diabetes onset in vivo. These results showed for the first time that CO interferes with T-cell priming by blocking an unknown mitochondria-dependent antigen-processing pathway in mature DC. Interestingly, other immune functions in DCs such as antigen capture, cytokine secretion, costimulation, and cell survival relied on glycolysis, suggesting that oxidative phosphorylation might only play a key role for the maturation of antigen-containing endosomes. In conclusion, CO produced by HO-1 impairs antigen-dependent inflammation by regulating DC immunogenicity by a mitochondria-dependent mechanism.


Subject(s)
Antigen Presentation/drug effects , Carbon Monoxide/pharmacology , Dendritic Cells/immunology , Endosomes/physiology , Mitochondria/physiology , Carbonyl Cyanide m-Chlorophenyl Hydrazone/pharmacology , Heme Oxygenase-1/physiology , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...