Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Children (Basel) ; 10(6)2023 May 24.
Article in English | MEDLINE | ID: mdl-37371160

ABSTRACT

Exergames are defined as digital games that require bodily movements to play, stimulating an active gaming experience to function as a form of physical activity (PA). The players interact with the game through whole-body movements improving energy expenditure. Exergames may be effective in improving physical and psychological aspects of children and adolescents with obesity. In this narrative review, we synthesized the current evidence regarding the role of exergames in modifying body composition and weight and in promoting changes in sedentary behavior to define the benefits of active video games as useful tools for fighting sedentarism and to outline the future directions of exergaming as a supplementation exercise rather than a replacement in educational programs for pediatric obesity. Data from the literature indicate that exergames may offer an interesting impact on childhood obesity and may be considered a potential strategy for controlling weight gain and body composition, promote PA, and decrease time spent on sedentary behavior in children and adolescents with obesity. However, exergame use also has some limits, such as children's poor self-regulation and poor structuring of exergame use. Therefore, a prudent approach should be maintained, and additional high-quality research is needed to determine if exergames can be effectively used in the treatment of childhood obesity and if new digital media, as a supplementation of exercise rather than a replacement, could be considered to combat sedentary behavior in educational programs for pediatric obesity prevention.

2.
Intensive Crit Care Nurs ; 70: 103186, 2022 Jun.
Article in English | MEDLINE | ID: mdl-34903466

ABSTRACT

BACKGROUND: Manual ventilation is life saving in critically ill patients. The lack of airway pressure monitoring makes it operator and device dependent. In this bench top-study, we compared a self- inflating bag valve resuscitator and a Mapleson C circuit during manual ventilation performed by critical care nurses under normal and pathologic conditions, with a special focus on delivered positive end expiratory pressure (PEEP). METHODS: Three different respiratory patterns (normal, restrictive and obstructive) were reproduced by a breathing simulator. Twenty nurses provided manual ventilation with a specific ventilatory pattern. Airway pressure, tidal volume and respiratory rate were recorded. Absolute value, error (difference between recorded and target values) and variability of PEEP were analysed. RESULTS: 3820 breathing traces were analysed. PEEP error was significantly higher with Mapelson C (43.3% vs 5.9% respectively, p < 0.001). This finding was confirmed regardless of operator skill and scenario. PEEP was more variable with Mapelson C (p < 0.05 in all scenarios). Ventilation of obstructive patients with Mapelson C resulted in higher PEEP levels compared to the reference value. Conversely, in the restrictive setting, PEEP was lower. Difference between PEEP and the minimum pressure recorded during the respiratory cycle was significantly higher with Mapelson C (p < 0.05). CONCLUSIONS: Manual ventilation with a Mapleson C circuit delivered a less accurate and less stable PEEP level compared to a self-inflating bag valve resuscitator.


Subject(s)
Respiration, Artificial , Resuscitation , Humans , Lung , Positive-Pressure Respiration , Respiration, Artificial/methods , Resuscitation/methods , Tidal Volume
3.
PLoS Pathog ; 17(8): e1009905, 2021 08.
Article in English | MEDLINE | ID: mdl-34449811

ABSTRACT

B1 cells, a subset of B lymphocytes whose developmental origin, phenotype, and function differ from that of conventional B2 cells, are the main source of "natural" IgM but can also respond to infection by rapidly producing pathogen-specific IgM directed against T-independent antigens. Francisella tularensis (Ft) is a Gram-negative bacterium that causes tularemia. Infection with Ft Live Vaccine Strain activates B1 cells for production of IgM directed against the bacterial LPS in a process incompletely understood. Here we show that immunization with purified Ft LPS elicits production of LPS-specific IgM and IgG3 by B1 cells independently of TLR2 or MyD88. Immunization, but not infection, generated peritoneum-resident memory B1 cells that differentiated into LPS-specific antibody secreting cells (ASC) upon secondary challenge. IL-5 was rapidly induced by immunization with Ft LPS and was required for production of LPS-specific IgM. Antibody-mediated depletion of ILC2 indicated that these cells were the source of IL-5 and were required for IgM production. IL-25, an alarmin that strongly activates ILC2, was rapidly secreted in response to immunization or infection and its administration to mice significantly increased IgM production and B1 cell differentiation to ASC. Conversely, mice lacking IL-17RB, the IL-25 receptor, showed impaired IL-5 induction, IgM production, and B1 ASC differentiation in response to immunization. Administration of IL-5 to Il17rb-/- mice rescued these B1 cells-mediated responses. Il17rb-/- mice were more susceptible to infection with Ft LVS and failed to develop immunity upon secondary challenge suggesting that LPS-specific IgM is one of the protective adaptive immune mechanisms against tularemia. Our results indicated that immunization with Ft LPS triggers production of IL-25 that, through stimulation of IL-5 release by ILC2, promotes B1 cells activation and differentiation into IgM secreting cells. By revealing the existence of an IL-25-ILC2-IL-5 axis our results suggest novel strategies to improve vaccination against T-independent bacterial antigens.


Subject(s)
Antibodies, Bacterial/immunology , B-Lymphocyte Subsets/immunology , Francisella tularensis/immunology , Immunoglobulin M/immunology , Interleukin-5/metabolism , Interleukins/metabolism , Lipopolysaccharides/pharmacology , Animals , Antibodies, Bacterial/metabolism , B-Lymphocyte Subsets/metabolism , Immunity, Innate , Immunoglobulin M/metabolism , Interleukin-5/genetics , Interleukins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/physiology , Receptors, Interleukin-17/physiology , Toll-Like Receptor 2/physiology , Tularemia/immunology , Tularemia/microbiology , Tularemia/pathology
4.
J Immunol ; 202(12): 3468-3473, 2019 06 15.
Article in English | MEDLINE | ID: mdl-31036765

ABSTRACT

Gasdermin D (GSDMD) cleavage by caspase-1 or caspase-11 inflammasomes triggers pyroptosis, a lytic form of cell death protective against intracellular bacteria. In this study, we examine the role of GSDMD in a mouse model of melioidosis. Gsdmd-/- mice were more susceptible than wild-type mice to intranasal infection with Burkholderia thailandensis Production of IL-18, but not IL-1ß, was decreased in Gsdmd-/- infected mice. Despite lower IL-18, IFN-γ was produced in similar amounts in wild-type and Gsdmd-/- mice. In vitro, secretion of both IL-1ß and IL-18 by macrophages or dendritic cells infected with B. thailandensis was dependent on GSDMD. Surprisingly, wild-type or GSDMD-deficient neutrophils secreted similar amounts of IL-1ß, suggesting these cells may be the source of the GSDMD-independent IL-1ß detected in vivo. Recombinant GSDMD was able to directly kill B. thailandensis in vitro upon processing by active caspase-1. Moreover, bacteria harvested from wild-type, but not Gsdmd-/- , macrophages were more susceptible to the microbicidal effect of hydrogen peroxide or human ß-defensin-3. Finally, we provide evidence that pyroptosis of in vitro infected macrophages is directly microbicidal. Taken together, these results indicate that the protective action of GSDMD in melioidosis is primarily due to induction of pyroptosis and direct killing of bacteria rather than production of cytokines.


Subject(s)
Burkholderia Infections/immunology , Burkholderia/physiology , Interleukin-18/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Macrophages/immunology , Melioidosis/immunology , Phosphate-Binding Proteins/metabolism , Animals , Caspase 1/genetics , Caspase 1/metabolism , Caspases, Initiator/genetics , Caspases, Initiator/metabolism , Cells, Cultured , Interleukin-1beta/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphate-Binding Proteins/genetics , Pyroptosis
5.
PLoS Pathog ; 14(5): e1007105, 2018 05.
Article in English | MEDLINE | ID: mdl-29791511

ABSTRACT

Infection with Burkholderia pseudomallei or B. thailandensis triggers activation of the NLRP3 and NLRC4 inflammasomes leading to release of IL-1ß and IL-18 and death of infected macrophages by pyroptosis, respectively. The non-canonical inflammasome composed of caspase-11 is also activated by these bacteria and provides protection through induction of pyroptosis. The recent generation of bona fide caspase-1-deficient mice allowed us to reexamine in a mouse model of pneumonic melioidosis the role of caspase-1 independently of caspase-11 (that was also absent in previously generated Casp1-/- mice). Mice lacking either caspase-1 or caspase-11 were significantly more susceptible than wild type mice to intranasal infection with B. thailandensis. Absence of caspase-1 completely abolished production of IL-1ß and IL-18 as well as pyroptosis of infected macrophages. In contrast, in mice lacking caspase-11 IL-1ß and IL-18 were produced at normal level and macrophages pyroptosis was only marginally affected. Adoptive transfer of bone marrow indicated that caspase-11 exerted its protective action both in myeloid cells and in radio-resistant cell types. B. thailandensis was shown to readily infect mouse lung epithelial cells triggering pyroptosis in a caspase-11-dependent way in vitro and in vivo. Importantly, we show that lung epithelial cells do not express inflammasomes components or caspase-1 suggesting that this cell type relies exclusively on caspase-11 for undergoing cell death in response to bacterial infection. Finally, we show that IL-18's protective action in melioidosis was completely dependent on its ability to induce IFNγ production. In turn, protection conferred by IFNγ against melioidosis was dependent on generation of ROS through the NADPH oxidase but independent of induction of caspase-11. Altogether, our results identify two non-redundant protective roles for caspase-1 and caspase-11 in melioidosis: Caspase-1 primarily controls pyroptosis of infected macrophages and production of IL-18. In contrast, caspase-11 mediates pyroptosis of infected lung epithelial cells.


Subject(s)
Caspase 1/physiology , Caspases/physiology , Interleukin-18/metabolism , Lung/cytology , Melioidosis/prevention & control , Pyroptosis/physiology , Animals , Burkholderia/physiology , Caspases, Initiator , Cell Line , Disease Models, Animal , Female , Interferon-gamma/physiology , Macrophages/microbiology , Macrophages/physiology , Male , Melioidosis/immunology , Mice , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism , Respiratory Mucosa/cytology
6.
Curr Top Microbiol Immunol ; 397: 199-214, 2016.
Article in English | MEDLINE | ID: mdl-27460811

ABSTRACT

Burkholderia pseudomallei is a Gram-negative flagellate bacterium that causes melioidosis, a disease endemic to Southeast Asia and other tropical regions. Following infection of macrophages and other non-phagocytic cell types, B. pseudomallei or B. thailandensis (a related species that causes disease in mice but not humans) are able to escape the phagosome and replicate in the host cell cytoplasm. Resistance to infection with Burkholderia is dependent on the Nlrp3 and Nlrc4 inflammasomes and the non-canonical caspase-11 inflammasome. Nlrc4 mediates protection through induction of pyroptosis in the early phase of infection. As the infection progresses and as IL-18-dependent IFNγ production increases, caspase-11-dependent pyroptosis acquires a preponderant protective role. Production of IL-1ß and IL-18 during infection is primarily mediated by Nlrp3. IL-18 is essential for survival because of its ability to induce IFNγ production, which in turn activates macrophage microbicidal functions and primes for caspase-11 expression. In contrast, during melioidosis, IL-1ß has deleterious effects due to excessive recruitment of neutrophils to the lung and consequent tissue damage.


Subject(s)
Burkholderia Infections/immunology , Burkholderia pseudomallei/physiology , Inflammasomes/immunology , Animals , Burkholderia Infections/microbiology , Burkholderia pseudomallei/genetics , Burkholderia pseudomallei/immunology , Humans , Inflammasomes/genetics , Macrophages/immunology , Neutrophils/immunology
7.
PLoS Pathog ; 11(3): e1004706, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25768794

ABSTRACT

The role of IL-1ß and IL-18 during lung infection with the gram-negative bacterium Francisella tularensis LVS has not been characterized in detail. Here, using a mouse model of pneumonic tularemia, we show that both cytokines are protective, but through different mechanisms. Il-18-/- mice quickly succumb to the infection and showed higher bacterial burden in organs and lower level of IFNγ in BALF and serum compared to wild type C57BL/6J mice. Administration of IFNγ rescued the survival of Il-18-/- mice, suggesting that their decreased resistance to tularemia is due to inability to produce IFNγ. In contrast, mice lacking IL-1 receptor or IL-1ß, but not IL-1α, appeared to control the infection in its early stages, but eventually succumbed. IFNγ administration had no effect on Il-1r1-/- mice survival. Rather, Il-1r1-/- mice were found to have significantly reduced titer of Ft LPS-specific IgM. The anti-Ft LPS IgM was generated in a IL-1ß-, TLR2-, and ASC-dependent fashion, promoted bacteria agglutination and phagocytosis, and was protective in passive immunization experiments. B1a B cells produced the anti-Ft LPS IgM and these cells were significantly decreased in the spleen and peritoneal cavity of infected Il-1b-/- mice, compared to C57BL/6J mice. Collectively, our results show that IL-1ß and IL-18 activate non-redundant protective responses against tularemia and identify an essential role for IL-1ß in the rapid generation of pathogen-specific IgM by B1a B cells.


Subject(s)
Antibodies, Bacterial/immunology , B-Lymphocyte Subsets/immunology , Immunoglobulin M/immunology , Interleukin-1beta/immunology , Tularemia/immunology , Animals , Blotting, Western , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Lipopolysaccharides/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Respiratory Tract Infections/immunology
8.
PLoS Pathog ; 10(8): e1004327, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25166912

ABSTRACT

Two distinct defense strategies can protect the host from infection: resistance is the ability to destroy the infectious agent, and tolerance is the ability to withstand infection by minimizing the negative impact it has on the host's health without directly affecting pathogen burden. Burkholderia pseudomallei is a Gram-negative bacterium that infects macrophages and causes melioidosis. We have recently shown that inflammasome-triggered pyroptosis and IL-18 are equally important for resistance to B. pseudomallei, whereas IL-1ß is deleterious. Here we show that the detrimental role of IL-1ß during infection with B. pseudomallei (and closely related B. thailandensis) is due to excessive recruitment of neutrophils to the lung and consequent tissue damage. Mice deficient in the potentially damaging enzyme neutrophil elastase were less susceptible than the wild type C57BL/6J mice to infection, although the bacterial burdens in organs and the extent of inflammation were comparable between C57BL/6J and elastase-deficient mice. In contrast, lung tissue damage and vascular leakage were drastically reduced in elastase-deficient mice compared to controls. Bradykinin levels were higher in C57BL/6 than in elastase-deficient mice; administration of a bradykinin antagonist protected mice from infection, suggesting that increased vascular permeability mediated by bradykinin is one of the mechanisms through which elastase decreases host tolerance to melioidosis. Collectively, these results demonstrate that absence of neutrophil elastase increases host tolerance, rather than resistance, to infection by minimizing host tissue damage.


Subject(s)
Burkholderia Infections/immunology , Host-Parasite Interactions/immunology , Leukocyte Elastase/immunology , Leukocyte Elastase/metabolism , Respiratory Tract Infections/immunology , Animals , Blotting, Western , Burkholderia Infections/enzymology , Disease Models, Animal , Flow Cytometry , Lung/microbiology , Lung/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Respiratory Tract Infections/enzymology , Respiratory Tract Infections/microbiology
9.
ScientificWorldJournal ; 11: 2037-50, 2011.
Article in English | MEDLINE | ID: mdl-22125454

ABSTRACT

The inflammasome is an important innate immune pathway that regulates at least two host responses protective against infections: (1) secretion of the proinflammatory cytokines IL-1ß and IL-18 and (2) induction of pyroptosis, a form of cell death. Inflammasomes, of which different types have been identified, are multiprotein complexes containing pattern recognition receptors belonging to the Nod-like receptor family or the PYHIN family and the protease caspase-1. The molecular aspects involved in the activation of different inflammasomes by various pathogens are being rapidly elucidated, and their role during infections is being characterized. Production of IL-1ß and IL-18 and induction of pyroptosis of the infected cell have been shown to be protective against many infectious agents. Here, we review the recent literature concerning inflammasome activation in the context of bacterial infections and identify important questions to be answered in the future.


Subject(s)
Bacterial Infections/immunology , Inflammasomes/physiology , Interleukin-18/physiology , Interleukin-1beta/physiology , Humans , Interleukin-18/immunology , Interleukin-1beta/immunology
10.
Infect Immun ; 79(12): 4828-38, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21947769

ABSTRACT

Mycobacterium tuberculosis is a highly successful human pathogen that evades host innate immunity by interfering with macrophage functions. In addition to avoiding macrophage microbicidal activities, M. tuberculosis triggers secretion of proinflammatory cytokines and chemokines in macrophages. The levels of proinflammatory cytokines induced by clinical M. tuberculosis isolates are thought to play an important role in determining tuberculosis disease progression and severity, but the mechanisms by which M. tuberculosis modulates the magnitude of inflammatory responses remain unclear. Here we show that M. tuberculosis restricts robust macrophage activation and dampens proinflammatory responses through the cell envelope-associated serine hydrolase Hip1 (hydrolase important for pathogenesis 1). By transcriptionally profiling macrophages infected with either wild-type or hip1 mutant bacteria, we found that the hip1 mutant induced earlier and significantly higher levels of several proinflammatory cytokines and chemokines. We show that increased activation of Toll-like receptor 2 (TLR2)- and MyD88-dependent signaling pathways mediates the enhanced cytokine secretion induced by the hip1 mutant. Thus, Hip1 restricts the onset and magnitude of proinflammatory cytokines by limiting TLR2-dependent activation. We also show that Hip1 dampens TLR2-independent activation of the inflammasome and limits secretion of interleukin-18 (IL-18). Dampening of TLR2 signaling does not require viable M. tuberculosis or phagocytosis but does require Hip1 catalytic activity. We propose that M. tuberculosis restricts proinflammatory responses by masking cell surface interactions between TLR2 agonists on M. tuberculosis and TLR2 on macrophages. This strategy may allow M. tuberculosis to evade early detection by host immunity, delay the onset of adaptive immune responses, and accelerate disease progression.


Subject(s)
Bacterial Proteins/metabolism , Gene Expression Regulation/drug effects , Macrophages/drug effects , Macrophages/physiology , Mycobacterium tuberculosis/metabolism , Serine Proteases/metabolism , Toll-Like Receptor 2/metabolism , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cells, Cultured , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein , Signal Transduction , Toll-Like Receptor 2/genetics
11.
BMC Microbiol ; 11: 179, 2011 Aug 05.
Article in English | MEDLINE | ID: mdl-21819572

ABSTRACT

BACKGROUND: A number of studies have revealed that Francisella tularensis (FT) suppresses innate immune responses such as chemokine/cytokine production and neutrophil recruitment in the lungs following pulmonary infection via an unidentified mechanism. The ability of FT to evade early innate immune responses could be a very important virulence mechanism for this highly infectious bacterial pathogen. RESULTS: Here we describe the characterization of a galU mutant strain of FT live vaccine strain (LVS). We show that the galU mutant was highly attenuated in a murine model of tularemia and elicited more robust innate immune responses than the wild-type (WT) strain. These studies document that the kinetics of chemokine expression and neutrophil recruitment into the lungs of mice challenged with the galU mutant strain are significantly more rapid than observed with WT FT, despite the fact that there were no observed differences in TLR2 or TLR4 signaling or replication/dissemination kinetics during the early stages of infection. We also show that the galU mutant had a hypercytotoxic phenotype and more rapidly induced the production of IL-1ß following infection either in vitro or in vivo, indicating that attenuation of the galU mutant strain may be due (in part) to more rapid activation of the inflammasome and/or earlier death of FT infected cells. Furthermore, we show that infection of mice with the galU mutant strain elicits protective immunity to subsequent challenge with WT FT. CONCLUSIONS: Disruption of the galU gene of FTLVS has little (if any) effect on in vivo infectivity, replication, or dissemination characteristics, but is highly attenuating for virulence. The attenuated phenotype of this mutant strain of FT appears to be related to its increased ability to induce innate inflammatory responsiveness, resulting in more rapid recruitment of neutrophils to the lungs following pneumonic infection, and/or to its ability to kill infected cells in an accelerated fashion. These results have identified two potentially important virulence mechanisms used by FT. These findings could also have implications for design of a live attenuated vaccine strain of FT because sublethal infection of mice with the galU mutant strain of FTLVS promoted development of protective immunity to WT FTLVS.


Subject(s)
Francisella tularensis/genetics , Francisella tularensis/pathogenicity , Tularemia/microbiology , Tularemia/pathology , UTP-Glucose-1-Phosphate Uridylyltransferase/deficiency , Virulence Factors/deficiency , Animals , Chemokines/metabolism , Disease Models, Animal , Francisella tularensis/immunology , Humans , Interleukin-1beta/immunology , Lung/immunology , Lung/microbiology , Mice , Mice, Inbred C57BL , Neutrophils/immunology , Pneumonia, Bacterial/microbiology , Pneumonia, Bacterial/pathology , Rodent Diseases/microbiology , Rodent Diseases/pathology , Toll-Like Receptor 2/immunology , Toll-Like Receptor 4/immunology , Virulence
12.
PLoS Pathog ; 7(12): e1002452, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22241982

ABSTRACT

Burkholderia pseudomallei is a Gram-negative bacterium that infects macrophages and other cell types and causes melioidosis. The interaction of B. pseudomallei with the inflammasome and the role of pyroptosis, IL-1ß, and IL-18 during melioidosis have not been investigated in detail. Here we show that the Nod-like receptors (NLR) NLRP3 and NLRC4 differentially regulate pyroptosis and production of IL-1ß and IL-18 and are critical for inflammasome-mediated resistance to melioidosis. In vitro production of IL-1ß by macrophages or dendritic cells infected with B. pseudomallei was dependent on NLRC4 and NLRP3 while pyroptosis required only NLRC4. Mice deficient in the inflammasome components ASC, caspase-1, NLRC4, and NLRP3, were dramatically more susceptible to lung infection with B. pseudomallei than WT mice. The heightened susceptibility of Nlrp3⁻/⁻ mice was due to decreased production of IL-18 and IL-1ß. In contrast, Nlrc4⁻/⁻ mice produced IL-1ß and IL-18 in higher amount than WT mice and their high susceptibility was due to decreased pyroptosis and consequently higher bacterial burdens. Analyses of IL-18-deficient mice revealed that IL-18 is essential for survival primarily because of its ability to induce IFNγ production. In contrast, studies using IL-1RI-deficient mice or WT mice treated with either IL-1ß or IL-1 receptor agonist revealed that IL-1ß has deleterious effects during melioidosis. The detrimental role of IL-1ß appeared to be due, in part, to excessive recruitment of neutrophils to the lung. Because neutrophils do not express NLRC4 and therefore fail to undergo pyroptosis, they may be permissive to B. pseudomallei intracellular growth. Administration of neutrophil-recruitment inhibitors IL-1ra or the CXCR2 neutrophil chemokine receptor antagonist antileukinate protected Nlrc4⁻/⁻ mice from lethal doses of B. pseudomallei and decreased systemic dissemination of bacteria. Thus, the NLRP3 and NLRC4 inflammasomes have non-redundant protective roles in melioidosis: NLRC4 regulates pyroptosis while NLRP3 regulates production of protective IL-18 and deleterious IL-1ß.


Subject(s)
Burkholderia pseudomallei/metabolism , Inflammasomes/metabolism , Interleukin-18/metabolism , Interleukin-1beta/metabolism , Melioidosis/metabolism , Pneumonia, Bacterial/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Humans , Inflammasomes/genetics , Interleukin-18/genetics , Interleukin-1beta/genetics , Melioidosis/genetics , Melioidosis/prevention & control , Mice , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Neutrophils/metabolism , Pneumonia, Bacterial/genetics , Pneumonia, Bacterial/prevention & control , Receptors, Interleukin-1 Type I/genetics , Receptors, Interleukin-1 Type I/metabolism , Receptors, Interleukin-8B/genetics , Receptors, Interleukin-8B/metabolism
13.
Mol Cell ; 39(3): 421-32, 2010 Aug 13.
Article in English | MEDLINE | ID: mdl-20705243

ABSTRACT

Cyclic phosphatidic acid (1-acyl-2,3-cyclic-glycerophosphate, CPA), one of nature's simplest phospholipids, is found in cells from slime mold to humans and has a largely unknown function. We find here that CPA is generated in mammalian cells in a stimulus-coupled manner by phospholipase D2 (PLD2) and binds to and inhibits the nuclear hormone receptor PPARgamma with nanomolar affinity and high specificity through stabilizing its interaction with the corepressor SMRT. CPA production inhibits the PPARgamma target-gene transcription that normally drives adipocytic differentiation of 3T3-L1 cells, lipid accumulation in RAW264.7 cells and primary mouse macrophages, and arterial wall remodeling in a rat model in vivo. Inhibition of PLD2 by shRNA, a dominant-negative mutant, or a small molecule inhibitor blocks CPA production and relieves PPARgamma inhibition. We conclude that CPA is a second messenger and a physiological inhibitor of PPARgamma, revealing that PPARgamma is regulated by endogenous agonists as well as by antagonists.


Subject(s)
Adipocytes/metabolism , Macrophages/metabolism , PPAR gamma/metabolism , Phosphatidic Acids/metabolism , Phospholipase D/metabolism , 3T3-L1 Cells , Animals , Cell Differentiation/physiology , Mice , Nuclear Receptor Co-Repressor 2/genetics , Nuclear Receptor Co-Repressor 2/metabolism , PPAR gamma/genetics , Phosphatidic Acids/genetics , Phospholipase D/genetics , Rats , Transcription, Genetic/physiology
14.
J Immunol ; 183(3): 1528-32, 2009 Aug 01.
Article in English | MEDLINE | ID: mdl-19596994

ABSTRACT

Cells undergoing necrosis release endogenous danger signals that possess proinflammatory potential. In this study we show that mature IL-1beta and IL-18 are released by necrotic cells but not by apoptotic cells. We identify 7-bromoindirubin-3'-oxime, an indirubin oxime derivative that induces necrosis, as a potent inducer of caspase-1 activation and release of mature IL-1beta and IL-18. Inflammasome activation was triggered by other necrosis-inducing treatments but was not observed in response to apoptosis-inducing stimuli. Necrosis-induced inflammasome activation was mediated by the NLRP3 and ASC molecules. Release of IL-18 and IL-1beta in response to necrosis-inducing stimuli was observed in THP-1 macrophages and the MSTO-211H human mesothelioma cell line independently of LPS priming. Using the in vivo model of naphthalene-induced airway epithelial cell injury, we showed that necrosis activates the ASC inflammasome in vivo. Our study identifies a new mechanism through which necrosis generates proinflammatory molecules that contributes to the sterile inflammatory response.


Subject(s)
Carrier Proteins/metabolism , Inflammation/etiology , Necrosis/pathology , Respiratory System/pathology , Animals , Cell Line , Epithelial Cells/pathology , Humans , Indoles , Inflammation/pathology , Interleukin-18 , Interleukin-1beta , Macrophages/pathology , Mesothelioma/pathology , Mice , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Oximes
15.
J Immunol ; 181(1): 17-21, 2008 Jul 01.
Article in English | MEDLINE | ID: mdl-18566365

ABSTRACT

Alum is the only adjuvant approved for routine use in humans, although the basis for its adjuvanticity remains poorly understood. We have recently shown that alum activates caspase-1 and induces secretion of mature IL-1beta and IL-18. In this study we show that, in human and mouse macrophages, alum-induced secretion of IL-1beta, IL-18, and IL-33 is mediated by the NLR (nucleotide-binding domain leucine-rich repeat-containing) protein NLRP3 and its adaptor ASC, but not by NLRC4. Other particulate adjuvants, such as QuilA and chitosan, induce inflammasome activation in a NLRP3-dependent fashion, suggesting that activation of the NLRP3-inflammasome may be a common mechanism of action of particulate adjuvants. Importantly, we demonstrate that Ag-specific Ab production elicited by vaccines that contain alum is significantly impaired in NLRP3-deficient mice. Our results demonstrate for the first time a role for the NLRP3-inflammasome during development of the immune response elicited by alum-enhanced vaccination and suggest that therapeutic intervention aimed at NLRP3 may improve adjuvant efficacy.


Subject(s)
Adjuvants, Immunologic/pharmacology , Aluminum Compounds/pharmacology , Aluminum Hydroxide/pharmacology , Carrier Proteins/immunology , Carrier Proteins/metabolism , Phosphates/pharmacology , Animals , Carrier Proteins/genetics , Cell Line , Cells, Cultured , Humans , Immune System/drug effects , Immune System/immunology , Inflammation/immunology , Mice , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , RNA Interference
16.
J Leukoc Biol ; 83(5): 1207-17, 2008 May.
Article in English | MEDLINE | ID: mdl-18285403

ABSTRACT

Hypersensitivity pneumonitis is an interstitial lung disease that is characterized by alveolitis, granuloma formation, and in some patients, fibrosis. Using the Saccharopolyspora rectivirgula animal model of Farmer's lung disease, our laboratory has demonstrated that neutrophils play a critical role in IFN-gamma production during the acute phase of the disease. As IFN-gamma is necessary for granuloma formation, it is important to identify the factors that lead to neutrophil recruitment during disease. To begin to identify the pattern recognition receptors (PRRs) that initiate chemokine production, leading to neutrophil recruitment following S. rectivirgula exposure, we examined the role of MyD88 and TLR2. Our results demonstrate that neutrophil recruitment, as measured by flow cytometry and the myeloperoxidase assay, was abolished in the absence of MyD88 following S. rectivirgula exposure. The decrease in neutrophil recruitment was likely a result of a significant decrease in production of neutrophil chemokines MIP-2 and keratinocyte-derived chemokine. These results suggest that S. rectivirgula interacts with PRRs that are upstream of the MyD88 pathway to initiate cytokine and chemokine production. In vitro studies suggest that S. rectivirgula can interact with TLR2, and stimulation of adherent cells from TLR2 knockout (KO) mice with S. rectivirgula resulted in a significant decrease in MIP-2 production. However, TLR2 KO mice did not have a reduction in neutrophil recruitment compared with wild-type mice following S. rectivirgula exposure. The results from our studies suggest that one or more PRR(s) upstream of MyD88 are necessary for neutrophil recruitment following S. rectivirgula exposure.


Subject(s)
Alveolitis, Extrinsic Allergic/physiopathology , Gram-Negative Bacterial Infections/physiopathology , Myeloid Differentiation Factor 88/physiology , Neutrophils/physiology , Alveolitis, Extrinsic Allergic/microbiology , Animals , Bronchoalveolar Lavage , Bronchoalveolar Lavage Fluid/cytology , Crosses, Genetic , Female , HeLa Cells , Humans , Lung/pathology , Lung/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Saccharopolyspora , Toll-Like Receptor 2/deficiency , Toll-Like Receptor 2/physiology
17.
J Biol Chem ; 283(7): 3751-60, 2008 Feb 15.
Article in English | MEDLINE | ID: mdl-18079113

ABSTRACT

The innate immune response to Francisella tularensis is primarily mediated by TLR2, though the bacterial products that stimulate this receptor remain unknown. Here we report the identification of two Francisella lipoproteins, TUL4 and FTT1103, which activate TLR2. We demonstrate that TUL4 and FTT1103 stimulate chemokine production in human and mouse cells in a TLR2-dependent way. Using an assay that relies on chimeric TLR proteins, we show that TUL4 and FTT1103 stimulate exclusively the TLR2/TLR1 heterodimer. Our results also show that yet unidentified Francisella proteins, possibly unlipi-dated, have the ability to stimulate the TLR2/TLR6 heterodimer. Through domain-exchange analysis, we determined that an extended region that comprises LRR 9-17 in the extra-cellular portion of TLR1 mediates response to Francisella lipoproteins and triacylated lipopeptide. Substitution of the corresponding LRR of TLR6 with the LRR derived from TLR1 enables TLR6 to recognize TUL4, FTT1103, and triacylated lipopeptide. This study identifies for the first time specific Fran-cisella products capable of stimulating a proinflammatory response and the cellular receptors they trigger.


Subject(s)
Francisella tularensis/metabolism , Lipoproteins/metabolism , Toll-Like Receptor 1/metabolism , Toll-Like Receptor 2/metabolism , Base Sequence , Cells, Cultured , DNA Primers , Dimerization , Humans , Lipoproteins/physiology , Signal Transduction , Toll-Like Receptor 1/chemistry , Toll-Like Receptor 2/chemistry
18.
J Leukoc Biol ; 83(2): 409-18, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17991761

ABSTRACT

The immunomodulatory properties of peptidoglycan (PGN), a constituent of the bacterial cell wall, have been studied extensively but with contrasting results. Recent studies have demonstrated that the TLR2-mediated inflammatory responses elicited by Gram-positive PGN preparations are in fact a result of contaminating lipoproteins and lipoteichoic acid that can be removed only through sophisticated extraction procedures. Here, we report that commercial preparations of Staphylococcus aureus or Streptococcus pyogenes PGN are contaminated with bacterial superantigens (SAg). The T cell-derived cytokines IL-17A and IL-17F were induced by PGN preparations but not by TLR agonists or nucleotide-binding and oligomerization domain-like receptor agonists in human PBMC. IL-17 induction by PGN preparations was sensitive to protease digestion and required TCR signaling. Bacterial SAg could be detected by immunoblot in the PGN preparations, and purified recombinant SAg were powerful inducers of IL-17. Finally, the PGN preparations stimulated proliferation and expansion of T cells bearing specific TCR V beta elements. Our results suggest that a large body of literature that relied on commercial PGN preparations to study inflammatory diseases, such as arthritis, where IL-17 also plays an important role, should be interpreted with caution and possibly revisited. Future studies aimed at characterizing the activities of PGN should use PGN preparations of proven purity.


Subject(s)
Antigens, Bacterial/analysis , Artifacts , Drug Contamination , Gene Expression Regulation/drug effects , Interleukin-17/biosynthesis , Peptidoglycan/analysis , Staphylococcus aureus/chemistry , Streptococcus pyogenes/chemistry , Superantigens/analysis , T-Lymphocyte Subsets/drug effects , Antigens, Bacterial/pharmacology , Cell Line , Gene Rearrangement, beta-Chain T-Cell Antigen Receptor , Humans , Interleukin-17/genetics , Lymphocyte Activation/drug effects , Lymphokines/biosynthesis , Lymphokines/genetics , NF-kappa B/metabolism , Peptidoglycan/isolation & purification , Receptors, Antigen, T-Cell, alpha-beta/analysis , Superantigens/pharmacology , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , Toll-Like Receptor 2/agonists , Toll-Like Receptor 2/physiology
19.
J Biol Chem ; 283(3): 1257-1266, 2008 Jan 18.
Article in English | MEDLINE | ID: mdl-17977838

ABSTRACT

Potent mammalian cell activation by Gram-negative bacterial endotoxin requires sequential protein-endotoxin and protein-protein interactions involving lipopolysaccharide-binding protein, CD14, MD-2, and Toll-like receptor 4 (TLR4). TLR4 activation requires simultaneous binding of MD-2 to endotoxin (E) and the ectodomain of TLR4. We now describe mutants of recombinant human MD-2 that bind TLR4 and react with E.CD14 but do not support cellular responsiveness to endotoxin. The mutants F121A/K122A MD-2 and Y131A/K132A MD-2 react with E.CD14 only when co-expressed with TLR4. Single mutants K122A and K132A each react with E.CD14 +/- TLR4 and promote TLR4-dependent cell activation by endotoxin suggesting that Phe(121) and Tyr(131) are needed for TLR4-independent transfer of endotoxin from CD14 to MD-2 and also needed for TLR4 activation by bound E.MD-2. The mutant F126A MD-2 reacts as well as wild-type MD-2 with E.CD14 +/- TLR4. E.MD-2(F126A) binds TLR4 with high affinity (K(d) approximately 200 pm) but does not activate TLR4 and instead acts as a potent TLR4 antagonist, inhibiting activation of HEK/TLR4 cells by wild-type E.MD-2. These findings reveal roles of Phe(121) and Tyr(131) in TLR4-independent interactions of human MD-2 with E.CD14 and, together with Phe(126), in activation of TLR4 by bound E.MD-2. These findings strongly suggest that the structural properties of E.MD-2, not E alone, determine agonist or antagonist effects on TLR4.


Subject(s)
Endotoxins/metabolism , Lymphocyte Antigen 96/chemistry , Lymphocyte Antigen 96/metabolism , Phenylalanine/metabolism , Toll-Like Receptor 4/metabolism , Tyrosine/metabolism , Cell Line , Culture Media , Endotoxins/pharmacology , Humans , Lipopolysaccharide Receptors/metabolism , Lipopolysaccharides/pharmacology , Mutant Proteins/metabolism , Protein Binding/drug effects , Protein Structure, Tertiary , Solubility/drug effects , Structure-Activity Relationship , Toll-Like Receptor 4/antagonists & inhibitors , Toll-Like Receptor 4/chemistry , Transfection , Tritium
20.
J Immunol ; 178(8): 5271-6, 2007 Apr 15.
Article in English | MEDLINE | ID: mdl-17404311

ABSTRACT

Aluminum hydroxide (Alum) is the only adjuvant approved for routine use in humans, although the basis for its adjuvanticity remains poorly understood. In this study, we show that Alum activates caspase-1 and induce secretion of mature IL-1beta and IL-18. Human PBMC or dendritic cells stimulated with pure TLR4 and TLR2 agonists released only traces of IL-1beta or IL-18, despite the fact that the IL-1beta mRNA was readily induced by both TLR agonists. In contrast, cells costimulated with TLR agonists plus Alum released large amount of IL-1beta and IL-18. Alum-induced IL-1beta and IL-18 production was not due to enhancement of TLR signaling but rather reflected caspase-1 activation and in mouse dendritic cells occurred in a MyD88-independent fashion. Secretion of other proinflammatory cytokines such as IL-8 was not affected by Alum treatments. However, TLR-induced production of IL-10 was increased and that of IFN-gamma-inducible protein decreased by Alum cotreatment. Considering the immunostimulatory activities of these cytokines and the ability of IL-1beta to act as adjuvant, our results suggest a mechanism for the adjuvanticity of Alum.


Subject(s)
Adjuvants, Immunologic/pharmacology , Aluminum Hydroxide/pharmacology , Caspase 1/metabolism , Interleukin-18/biosynthesis , Interleukin-1beta/biosynthesis , Chemokine CXCL10 , Chemokines, CXC/biosynthesis , Enzyme Activation/drug effects , Humans , Interleukin-10/biosynthesis , Myeloid Differentiation Factor 88/physiology , Signal Transduction , Toll-Like Receptors/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...