Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
1.
Proc Natl Acad Sci U S A ; 119(21): e2202012119, 2022 05 24.
Article in English | MEDLINE | ID: mdl-35588457

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS­CoV-2) is a worldwide health concern, and new treatment strategies are needed. Targeting inflammatory innate immunity pathways holds therapeutic promise, but effective molecular targets remain elusive. Here, we show that human caspase-4 (CASP4) and its mouse homolog, caspase-11 (CASP11), are up-regulated in SARS­CoV-2 infections and that CASP4 expression correlates with severity of SARS­CoV-2 infection in humans. SARS­CoV-2­infected Casp11−/− mice were protected from severe weight loss and lung pathology, including blood vessel damage, compared to wild-type (WT) mice and mice lacking the caspase downstream effector gasdermin-D (Gsdmd−/−). Notably, viral titers were similar regardless of CASP11 knockout. Global transcriptomics of SARS­CoV-2­infected WT, Casp11−/−, and Gsdmd−/− lungs identified restrained expression of inflammatory molecules and altered neutrophil gene signatures in Casp11−/− mice. We confirmed that protein levels of inflammatory mediators interleukin (IL)-1ß, IL-6, and CXCL1, as well as neutrophil functions, were reduced in Casp11−/− lungs. Additionally, Casp11−/− lungs accumulated less von Willebrand factor, a marker for endothelial damage, but expressed more Kruppel-Like Factor 2, a transcription factor that maintains vascular integrity. Overall, our results demonstrate that CASP4/11 promotes detrimental SARS­CoV-2­induced inflammation and coagulopathy, largely independently of GSDMD, identifying CASP4/11 as a promising drug target for treatment and prevention of severe COVID-19.


Subject(s)
COVID-19 , Caspases, Initiator/metabolism , SARS-CoV-2 , Thromboinflammation , Animals , COVID-19/enzymology , COVID-19/pathology , Caspases, Initiator/genetics , Disease Progression , Humans , Lung/pathology , Mice , Mice, Knockout , Severity of Illness Index , Thromboinflammation/enzymology , Thromboinflammation/genetics
2.
J Biol Chem ; 298(6): 102024, 2022 06.
Article in English | MEDLINE | ID: mdl-35568196

ABSTRACT

Apoptosis-associated speck-like protein containing a caspase recruitment domain (CARD) (ASC) is a 22 kDa protein that functions as the central adaptor for inflammasome assembly. ASC forms insoluble specks in monocytes undergoing pyroptosis, and the polymerization of ASC provides a template of CARDs that leads to proximity-mediated autoactivation of caspase-1 in canonical inflammasomes. However, specks are insoluble protein complexes, and solubility is typically important for protein function. Therefore, we sought to define whether ASC specks comprise active inflammasome complexes or are simply the end stage of exhausted ASC polymers. Using a THP-1 cell-lysing model of caspase-1 activation that is ASC dependent, we compared caspase-1 activation induced by preassembled insoluble ASC specks and soluble monomeric forms of ASC. Unexpectedly, after controlling for the concentration dependence of ASC oligomerization, we found that only insoluble forms of ASC promoted caspase-1 autocatalysis. This link to insolubility was recapitulated with recombinant ASC. We show that purified recombinant ASC spontaneously precipitated and was functional, whereas the maltose-binding protein-ASC fusion to ASC (promoting enhanced solubility) was inactive until induced to insolubility by binding to amylose beads. This functional link to insolubility also held true for the Y146A mutation of the CARD of ASC, which avoids insolubility and caspase-1 activation. Thus, we conclude that the role of ASC insolubility in inflammasome function is inextricably linked to its pyrin domain-mediated and CARD-mediated polymerizations. These findings will support future studies into the molecular mechanisms controlling ASC solubility.


Subject(s)
CARD Signaling Adaptor Proteins , Caspase 1 , Inflammasomes , Adaptor Proteins, Signal Transducing/metabolism , CARD Signaling Adaptor Proteins/metabolism , Caspase 1/metabolism , Humans , Inflammasomes/metabolism , Pyroptosis , THP-1 Cells
3.
J Immunol ; 206(6): 1329-1336, 2021 03 15.
Article in English | MEDLINE | ID: mdl-33568399

ABSTRACT

Inflammasome activation is regulated in part by the posttranslational modification of inflammasome proteins. Tyrosine phosphorylation is one possible modification. Having previously shown that the protein tyrosine kinase (PTK) inhibitor AG126 greatly inhibits inflammasome activation, we sought to uncover the target kinase. To do this, we screened a commercial tyrosine kinase library for inhibition of inflammasome-dependent IL-18/IL-1ß release and pyroptosis. THP-1 cells (human monocyte cell line) were incubated with PTK inhibitors (0.1, 1, and 10 µM) before stimulation with LPS followed by ATP. The PTK inhibitors DCC-2036 (Rebastinib) and GZD824, specific for Bcr-Abl kinase, showed the most severe reduction of IL-18 and lactate dehydrogenase release at all concentrations used. The suggested kinase target, cAbl kinase, was then deleted in THP-1 cells by CRISPR/Cas9 editing and then tested for its role in inflammasome function and potential to phosphorylate the inflammasome adaptor ASC. The cABL knockout not only significantly inhibited inflammasome function but also decreased release of phosphorylated ASC after LPS/ATP stimulation. One predicted target of cAbl kinase is tyrosine 146 in ASC. Complementation of ASC knockout THP-1 cells with mutated Y146A ASC significantly abrogated inflammasome activation and ASC oligomerization as compared with wild-type ASC complementation. Thus, these findings support cAbl kinase as a positive regulator of inflammasome activity and pyroptosis, likely via phosphorylation of ASC.


Subject(s)
CARD Signaling Adaptor Proteins/metabolism , Inflammasomes/immunology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-abl/metabolism , Pyroptosis/immunology , Adenosine Triphosphate/immunology , Benzamides/pharmacology , CARD Signaling Adaptor Proteins/genetics , Gene Knock-In Techniques , Gene Knockout Techniques , Humans , Inflammasomes/drug effects , Inflammasomes/metabolism , Lipopolysaccharides/immunology , Mutation , Phosphorylation/drug effects , Phosphorylation/genetics , Phosphorylation/immunology , Proto-Oncogene Proteins c-abl/genetics , Pyrazoles/pharmacology , Pyridines/pharmacology , Pyroptosis/drug effects , Quinolines/pharmacology , THP-1 Cells , Tyrphostins/pharmacology
4.
Cell Immunol ; 370: 104425, 2021 12.
Article in English | MEDLINE | ID: mdl-34800762

ABSTRACT

Asthma is an inflammatory lung disorder characterized by mucus hypersecretion, cellular infiltration, and bronchial hyper-responsiveness. House dust mites (HDM) are the most prevalent cause of allergic sensitization. Canonical and noncanonical inflammasomes are multiprotein complexes that assemble in response to pathogen or danger-associated molecular patterns (PAMPs or DAMPs). Murine caspase-11 engages the noncanonical inflammasome. We addressed the role of caspase-11 in mediating host responses to HDM and subsequent allergic inflammation using caspase-11-/- mice, which lack caspase-11 while express caspase-1. We found that HDM induce caspase-11 expression in vitro. The presence of IL-4 and IL-13 promote caspase-11 expression. Additionally, caspase-11-/- macrophages show reduced release of IL-6, IL-12, and KC, and express lower levels of costimulatory molecules (e.g., CD40, CD86 and MHCII) in response to HDM stimulation. Notably, HDM sensitization of caspase-11-/- mice resulted in similar levels of IgE responses and hypothermia in response to nasal HDM challenge compared to WT. However, analysis of cell numbers and cytokines in bronchiolar alveolar lavage fluid (BALF) and histopathology of representative lung segments demonstrate altered inflammatory responses and reduced neutrophilia in the airways of the caspase-11-/- mice. These findings indicate that caspase-11 regulates airway inflammation in response to HDM exposure.


Subject(s)
Caspases, Initiator/immunology , Hypersensitivity/immunology , Pneumonia/immunology , Pyroglyphidae/immunology , Animals , Mice , Mice, Inbred C57BL , Mice, Knockout
5.
Immunity ; 37(1): 35-47, 2012 Jul 27.
Article in English | MEDLINE | ID: mdl-22658523

ABSTRACT

Inflammasomes are multiprotein complexes that include members of the NLR (nucleotide-binding domain leucine-rich repeat containing) family and caspase-1. Once bacterial molecules are sensed within the macrophage, the inflammasome is assembled, mediating the activation of caspase-1. Caspase-11 mediates caspase-1 activation in response to lipopolysaccharide and bacterial toxins, and yet its role during bacterial infection is unknown. Here, we demonstrated that caspase-11 was dispensable for caspase-1 activation in response to Legionella, Salmonella, Francisella, and Listeria. We also determined that active mouse caspase-11 was required for restriction of L. pneumophila infection. Similarly, human caspase-4 and caspase-5, homologs of mouse caspase-11, cooperated to restrict L. pneumophila infection in human macrophages. Caspase-11 promoted the fusion of the L. pneumophila vacuole with lysosomes by modulating actin polymerization through cofilin. However, caspase-11 was dispensable for the fusion of lysosomes with phagosomes containing nonpathogenic bacteria, uncovering a fundamental difference in the trafficking of phagosomes according to their cargo.


Subject(s)
Actins/metabolism , Bacteria/immunology , Caspases/metabolism , Lysosomes/metabolism , Phagosomes/metabolism , Protein Multimerization , Actin Depolymerizing Factors/metabolism , Animals , Bacteria/growth & development , Bacterial Infections/immunology , Bacterial Infections/metabolism , Caspase 1/deficiency , Caspase 1/genetics , Caspase 1/metabolism , Caspases/deficiency , Caspases/genetics , Caspases, Initiator , Humans , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phagosomes/microbiology , Phosphorylation
6.
EMBO Rep ; 20(12): e48109, 2019 12 05.
Article in English | MEDLINE | ID: mdl-31637841

ABSTRACT

Methicillin-resistant Staphylococcus aureus (MRSA) is a growing health concern due to increasing resistance to antibiotics. As a facultative intracellular pathogen, MRSA is capable of persisting within professional phagocytes including macrophages. Here, we identify a role for CASP11 in facilitating MRSA survival within murine macrophages. We show that MRSA actively prevents the recruitment of mitochondria to the vicinity of the vacuoles they reside in to avoid intracellular demise. This process requires CASP11 since its deficiency allows increased association of MRSA-containing vacuoles with mitochondria. The induction of mitochondrial superoxide by antimycin A (Ant A) improves MRSA eradication in casp11-/- cells, where mitochondria remain in the vicinity of the bacterium. In WT macrophages, Ant A does not affect MRSA persistence. When mitochondrial dissociation is prevented by the actin depolymerizing agent cytochalasin D, Ant A effectively reduces MRSA numbers. Moreover, the absence of CASP11 leads to reduced cleavage of CASP1, IL-1ß, and CASP7, as well as to reduced production of CXCL1/KC. Our study provides a new role for CASP11 in promoting the persistence of Gram-positive bacteria.


Subject(s)
Caspases, Initiator/metabolism , Macrophages/immunology , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Staphylococcal Infections/immunology , Animals , Anti-Bacterial Agents/pharmacology , Antimycin A/pharmacology , Caspases, Initiator/genetics , Cells, Cultured , Macrophages/microbiology , Methicillin-Resistant Staphylococcus aureus/drug effects , Mice , Mice, Inbred C57BL , Staphylococcal Infections/microbiology , Vacuoles/metabolism
7.
Immunity ; 32(6): 803-14, 2010 Jun 25.
Article in English | MEDLINE | ID: mdl-20620944

ABSTRACT

Among human natural killer (NK) cell intermediates in secondary lymphoid tissue (SLT), stage 3 CD34(-)CD117(+)CD161(+)CD94(-) immature NK (iNK) cells uniquely express aryl hydrocarbon receptor (AHR) and interleukin-22 (IL-22), supporting a role in mucosal immunity. The mechanisms controlling proliferation and differentiation of these cells are unknown. Here we demonstrate that the IL-1 receptor IL-1R1 was selectively expressed by a subpopulation of iNK cells that localized proximal to IL-1beta-producing conventional dendritic cells (cDCs) within SLT. IL-1R1(hi) iNK cells required continuous exposure to IL-1beta to retain AHR and IL-22 expression, and they proliferate in direct response to cDC-derived IL-15 and IL-1beta. In the absence of IL-1beta, a substantially greater fraction of IL-1R1(hi) iNK cells differentiated to stage 4 NK cells and acquired the ability to kill and secrete IFN-gamma. Thus, cDC-derived IL-1beta preserves and expands IL-1R1(hi)IL-22(+)AHR(+) iNK cells, potentially influencing human mucosal innate immunity during infection.


Subject(s)
Cell Differentiation/immunology , Interleukin-1beta/immunology , Interleukins/immunology , Killer Cells, Natural/cytology , Cell Proliferation , Cell Separation , Dendritic Cells/immunology , Dendritic Cells/metabolism , Flow Cytometry , Humans , Immunity, Mucosal/immunology , Immunohistochemistry , Interleukin-1beta/metabolism , Interleukins/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lymphoid Tissue/cytology , Lymphoid Tissue/immunology , Receptors, Interleukin-1/immunology , Receptors, Interleukin-1/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Interleukin-22
9.
Am J Respir Cell Mol Biol ; 59(1): 56-64, 2018 07.
Article in English | MEDLINE | ID: mdl-29365280

ABSTRACT

Lung endothelial cell apoptosis and injury occur throughout all stages of acute lung injury/acute respiratory distress syndrome and impact disease progression. Caspases 1, 4, and 5 are essential for completion of the apoptotic program known as pyroptosis that also involves proinflammatory cytokines. Because gasdermin D (GSDMD) mediates pyroptotic death and is essential for pore formation, we hypothesized that it might direct caspase 1-encapsulated microparticle (MP) release and mediate endothelial cell death. Our present work provides evidence that GSDMD is released by LPS-stimulated THP-1 monocytic cells, where it is packaged into microparticles together with active caspase 1. Furthermore, only MP released from stimulated monocytic cells that contain both cleaved GSDMD and active caspase 1 induce endothelial cell apoptosis. MPs pretreated with caspase 1 inhibitor Y-VAD or pan-caspase inhibitor Z-VAD do not contain cleaved GSDMD. MPs from caspase 1-knockout cells are also deficient in p30 active GSDMD, further confirming that caspase 1 regulates GSDMD function. Although control MPs contained cleaved GSDMD without caspase 1, these fractions were unable to induce cell death, suggesting that encapsulation of both caspase 1 and GSDMD is essential for cell death induction. Release of microparticulate active caspase 1 was abrogated in GSDMD knockout cells, although cytosolic caspase 1 activation was not impaired. Last, higher concentrations of microparticulate GSDMD were detected in the plasma of septic patients with acute respiratory distress syndrome than in that of healthy donors. Taken together, these findings suggest that GSDMD regulates the release of microparticulate active caspase 1 from monocytes essential for induction of cell death and thereby may play a critical role in sepsis-induced endothelial cell injury.


Subject(s)
Caspase 1/metabolism , Cell-Derived Microparticles/metabolism , Endothelial Cells/pathology , Lung Injury/pathology , Neoplasm Proteins/metabolism , Endothelial Cells/metabolism , Humans , Intracellular Signaling Peptides and Proteins , Lipopolysaccharides/pharmacology , Lung/pathology , Lung Injury/metabolism , Middle Aged , Phosphate-Binding Proteins , Sepsis/blood , Sepsis/pathology , THP-1 Cells
10.
Infect Immun ; 86(10)2018 10.
Article in English | MEDLINE | ID: mdl-30061379

ABSTRACT

Listeria monocytogenes is a facultative intracellular pathogen that infects a wide variety of cells, causing the life-threatening disease listeriosis. L. monocytogenes virulence factors include two surface invasins, InlA and InlB, known to promote bacterial uptake by host cells, and the secreted pore-forming toxin listeriolysin O (LLO), which disrupts the phagosome to allow bacterial proliferation in the cytosol. In addition, plasma membrane perforation by LLO has been shown to facilitate L. monocytogenes internalization into epithelial cells. In this work, we tested the host cell range and importance of LLO-mediated L. monocytogenes internalization relative to the canonical invasins, InlA and InlB. We measured the efficiencies of L. monocytogenes association with and internalization into several human cell types (hepatocytes, cytotrophoblasts, and endothelial cells) using wild-type bacteria and isogenic single, double, and triple deletion mutants for the genes encoding InlA, InlB and LLO. No role for InlB was detected in any tested cells unless the InlB expression level was substantially enhanced, which was achieved by introducing a mutation (prfA*) in the gene encoding the transcription factor PrfA. In contrast, InlA and LLO were the most critical invasion factors, although they act in a different manner and in a cell-type-dependent fashion. As expected, InlA facilitates both bacterial attachment and internalization in cells that express its receptor, E-cadherin. LLO promotes L. monocytogenes internalization into hepatocytes, but not into cytotrophoblasts and endothelial cells. Finally, LLO and InlA cooperate to increase the efficiency of host cell invasion by L. monocytogenes.


Subject(s)
Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Heat-Shock Proteins/metabolism , Hemolysin Proteins/metabolism , Listeria monocytogenes/metabolism , Listeriosis/microbiology , Membrane Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Toxins/genetics , Cadherins/genetics , Cadherins/metabolism , Heat-Shock Proteins/genetics , Hemolysin Proteins/genetics , Hepatocytes/metabolism , Hepatocytes/microbiology , Humans , Listeria monocytogenes/genetics , Listeria monocytogenes/pathogenicity , Listeriosis/genetics , Listeriosis/metabolism , Membrane Proteins/genetics , Peptide Termination Factors/genetics , Peptide Termination Factors/metabolism , Virulence
11.
PLoS Pathog ; 12(12): e1006035, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27911947

ABSTRACT

Type III secretion systems (T3SS) are central virulence factors for many pathogenic Gram-negative bacteria, and secreted T3SS effectors can block key aspects of host cell signaling. To counter this, innate immune responses can also sense some T3SS components to initiate anti-bacterial mechanisms. The Yersinia pestis T3SS is particularly effective and sophisticated in manipulating the production of pro-inflammatory cytokines IL-1ß and IL-18, which are typically processed into their mature forms by active caspase-1 following inflammasome formation. Some effectors, like Y. pestis YopM, may block inflammasome activation. Here we show that YopM prevents Y. pestis induced activation of the Pyrin inflammasome induced by the RhoA-inhibiting effector YopE, which is a GTPase activating protein. YopM blocks YopE-induced Pyrin-mediated caspase-1 dependent IL-1ß/IL-18 production and cell death. We also detected YopM in a complex with Pyrin and kinases RSK1 and PKN1, putative negative regulators of Pyrin. In contrast to wild-type mice, Pyrin deficient mice were also highly susceptible to an attenuated Y. pestis strain lacking YopM, emphasizing the importance of inhibition of Pyrin in vivo. A complex interplay between the Y. pestis T3SS and IL-1ß/IL-18 production is evident, involving at least four inflammasome pathways. The secreted effector YopJ triggers caspase-8- dependent IL-1ß activation, even when YopM is present. Additionally, the presence of the T3SS needle/translocon activates NLRP3 and NLRC4-dependent IL-1ß generation, which is blocked by YopK, but not by YopM. Taken together, the data suggest YopM specificity for obstructing the Pyrin pathway, as the effector does not appear to block Y. pestis-induced NLRP3, NLRC4 or caspase-8 dependent caspase-1 processing. Thus, we identify Y. pestis YopM as a microbial inhibitor of the Pyrin inflammasome. The fact that so many of the Y. pestis T3SS components are participating in regulation of IL-1ß/IL-18 release suggests that these effects are essential for maximal control of innate immunity during plague.


Subject(s)
Bacterial Outer Membrane Proteins/immunology , Inflammasomes/immunology , Plague/immunology , Pyrin/immunology , Animals , Disease Models, Animal , Mice , Mice, Knockout , Yersinia pestis/immunology
12.
J Immunol ; 197(4): 1322-34, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27421477

ABSTRACT

Immunosuppression is a major complication of alcoholism that contributes to increased rates of opportunistic infections and sepsis in alcoholics. The NLRP3 inflammasome, a multiprotein intracellular pattern recognition receptor complex that facilitates the cleavage and secretion of the proinflammatory cytokines IL-1ß and IL-18, can be inhibited by ethanol, and we sought to better understand the mechanism through which this occurs and whether chemically similar molecules exert comparable effects. We show that ethanol can specifically inhibit activation of the NLRP3 inflammasome, resulting in attenuated IL-1ß and caspase-1 cleavage and secretion, as well as diminished apoptosis-associated speck-like protein containing a CARD (ASC) speck formation, without affecting potassium efflux, in a mouse macrophage cell line (J774), mouse bone marrow-derived dendritic cells, mouse neutrophils, and human PBMCs. The inhibitory effects on the Nlrp3 inflammasome were independent of γ-aminobutyric acid A receptor activation or N-methyl-d-asparate receptor inhibition but were associated with decreased oxidant production. Ethanol treatment markedly decreased cellular tyrosine phosphorylation, whereas administration of the tyrosine phosphatase inhibitor sodium orthovanadate prior to ethanol restored tyrosine phosphorylation and IL-1ß secretion subsequent to ATP stimulation. Furthermore, sodium orthovanadate-induced phosphorylation of ASC Y144, necessary and sufficient for Nlrp3 inflammasome activation, and secretion of phosphorylated ASC were inhibited by ethanol. Finally, multiple alcohol-containing organic compounds exerted inhibitory effects on the Nlrp3 inflammasome, whereas 2-methylbutane (isopentane), the analogous alkane of the potent inhibitor isoamyl alcohol (isopentanol), did not. Our results demonstrate that ethanol antagonizes the NLRP3 inflammasome at an apical event in its activation through the stimulation of protein tyrosine phosphatases, an effect shared by other short-chain alcohols.


Subject(s)
Alcohols/toxicity , Ethanol/toxicity , Inflammasomes/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/drug effects , Protein Tyrosine Phosphatases/drug effects , Animals , Blotting, Western , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Humans , Inflammasomes/metabolism , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Protein Tyrosine Phosphatases/metabolism
13.
J Immunol ; 192(8): 3881-8, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24623131

ABSTRACT

Caspase-1 activation is a central event in innate immune responses to many pathogenic infections and tissue damage. The NLRP3 inflammasome, a multiprotein scaffolding complex that assembles in response to two distinct steps, priming and activation, is required for caspase-1 activation. However, the detailed mechanisms of these steps remain poorly characterized. To investigate the process of LPS-mediated NLRP3 inflammasome priming, we used constitutively present pro-IL-18 as the caspase-1-specific substrate to allow study of the early events. We analyzed human monocyte caspase-1 activity in response to LPS priming, followed by activation with ATP. Within minutes of endotoxin priming, the NLRP3 inflammasome is licensed for ATP-induced release of processed IL-18, apoptosis-associated speck-forming complex containing CARD, and active caspase-1, independent of new mRNA or protein synthesis. Moreover, extracellular signal-regulated kinase 1 (ERK1) phosphorylation is central to the priming process. ERK inhibition and small interfering RNA-mediated ERK1 knockdown profoundly impair priming. In addition, proteasome inhibition prevents ERK phosphorylation and blocks priming. Scavenging reactive oxygen species with diphenylene iodonium also blocks both priming and ERK phosphorylation. These findings suggest that ERK1-mediated posttranslational modifications license the NLRP3 inflammasome to respond to the second signal ATP by inducing posttranslational events that are independent of new production of pro-IL-1ß and NOD-like receptor components.


Subject(s)
Inflammasomes , Lipopolysaccharides/immunology , MAP Kinase Signaling System , Proteasome Endopeptidase Complex/metabolism , Carrier Proteins/metabolism , Caspase 1/metabolism , Gene Expression Regulation/drug effects , Humans , Immunity, Innate , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Interleukin-18/metabolism , Interleukin-1beta/metabolism , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Models, Biological , Monocytes/immunology , Monocytes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein , Oxidants/pharmacology , Protein Kinase Inhibitors/pharmacology
14.
Am J Respir Cell Mol Biol ; 53(3): 400-11, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25629767

ABSTRACT

Asthma is a chronic lung disease characterized by inflammation centered upon bronchial epithelium. House dust mite is one of the most common respiratory allergens that trigger exacerbations of asthma. IκBζ (gene NFKBIZ) is a recently recognized member of the NF-κB family that can be induced in mononuclear phagocytes and lung epithelial cells and has been shown to play a prominent role in epithelial cell function. We therefore analyzed the role of IκBζ in regulating lung epithelial cell cytokine responses to house dust mite mix (HDM). We found that human bronchial epithelial cells express IκBζ and release IL-6 and granulocyte macrophage colony-stimulating factor (GMCSF) when cocultured with human monocytes and HDM. This response is blocked in the presence of IL-1 receptor antagonist (IL-1Ra), indicating that it is IL-1 mediated. Neither HDM-stimulated macrophages nor dendritic cells release IL-1ß and subsequently induce cytokine release from the bronchial epithelial cells. Rhodobacter sphaeroides LPS (RS-LPS), a TLR4 antagonist, blocks the ability of HDM to induce IκBζ and release GMCSF from epithelial cells cocultured with monocytes. Additionally, human bronchial epithelial cells show no induction of IκBζ or cytokine responses to direct HDM stimulation. Finally, NFKBIZ small interfering RNA-mediated knockdown in the bronchial epithelial cells suppresses the release of IL-1-induced IL-6 and GMCSF. Our findings indicate a possible role for monocyte recruitment and lung epithelial cell IκBζ in mediating asthma associated inflammation. Thus, IκBζ, IL-1Ra, and RS-LPS deserve future study as potential modulators of house dust mite-induced asthma.


Subject(s)
Allergens/immunology , Alveolar Epithelial Cells/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , I-kappa B Proteins/physiology , Interleukin-1beta/biosynthesis , Nuclear Proteins/physiology , Pyroglyphidae/immunology , Adaptor Proteins, Signal Transducing , Alveolar Epithelial Cells/immunology , Animals , Asthma/immunology , Asthma/metabolism , Cell Line , Cell Nucleus/metabolism , Coculture Techniques , Humans , Lipopolysaccharides/pharmacology , Monocytes/metabolism , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism
15.
Biochem Biophys Res Commun ; 465(2): 232-8, 2015 Sep 18.
Article in English | MEDLINE | ID: mdl-26253471

ABSTRACT

A chemiluminescent method is proposed for quantitation of NO generation in cell cultures. The method is based on activation of soluble guanylyl cyclase by NO. The product of the guanylyl cyclase reaction, pyrophosphate, is converted to ATP by ATP sulfurylase and ATP is detected in a luciferin-luciferase system. The method has been applied to the measurement of NO generated by activated murine macrophages (RAW 264.7) and bovine aortic endothelial cells. For macrophages activated by lipopolysaccharide and γ-interferon, the rate of NO production is about 100 amol/(cell·min). The rate was confirmed by the measurements of nitrite, the product of NO oxidation. For endothelial cells, the basal rate of NO generation is 5 amol/(cell·min); the rate approximately doubles upon activation by bradykinin, Ca(2+) ionophore A23187 or mechanical stress. For both types of cells the measured rate of NO generation is strongly affected by inhibitors of NO synthase. The sensitivity of the method is about 50 pM/min, allowing the registration of NO generated by 10(2)-10(4) cells. The enzyme-linked chemiluminescent method is two orders of magnitude more sensitive than fluorescent detection using 4-amino-5-methylamino-2',7'-difluorofluorescein (DAF-FM).


Subject(s)
Biological Assay , Endothelial Cells/metabolism , Firefly Luciferin/chemistry , Luciferases/chemistry , Luminescent Measurements/standards , Macrophages/metabolism , Nitric Oxide/analysis , Animals , Aorta/cytology , Aorta/drug effects , Aorta/metabolism , Bradykinin/pharmacology , Cattle , Cell Line , Cyclic GMP/metabolism , Diphosphates/chemistry , Diphosphates/metabolism , Endothelial Cells/cytology , Endothelial Cells/drug effects , Firefly Luciferin/metabolism , Guanosine Triphosphate/metabolism , Guanylate Cyclase/metabolism , Lipopolysaccharides/pharmacology , Luciferases/metabolism , Luminescence , Macrophages/cytology , Macrophages/drug effects , Mice , Nitric Oxide/biosynthesis , Nitric Oxide Donors/metabolism , Nitric Oxide Donors/pharmacology , Nitrites/chemistry , Nitrites/metabolism , Nitroso Compounds/metabolism , Nitroso Compounds/pharmacology , Receptors, Cytoplasmic and Nuclear/metabolism , Sensitivity and Specificity , Soluble Guanylyl Cyclase , Sulfate Adenylyltransferase/metabolism
16.
Transfusion ; 55(8): 1937-45, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25819532

ABSTRACT

BACKGROUND: We have previously shown that critically ill children transfused with red blood cells (RBCs) of longer storage durations have more suppressed monocyte function after transfusion compared to children transfused with fresher RBCs and that older stored RBCs directly suppress monocyte function in vitro, through unknown mechanisms. We hypothesized that RBC-derived microvesicles (MVs) were responsible for monocyte suppression. STUDY DESIGN AND METHODS: To determine the role of stored RBC unit-derived MVs, we cocultured monocytes with supernatants, isolated MVs, or supernatants that had been depleted of MVs from prestorage leukoreduced RBCs that had been stored for either 7 or 30 days. Isolated MVs were characterized by electron microscopy and flow cytometry. Monocyte function after coculture experiments was measured by cytokine production after stimulation with lipopolysaccharide (LPS). RESULTS: Monocyte function was suppressed after exposure to supernatants from 30-day RBC units compared to monocytes cultured in medium alone (LPS-induced tumor necrosis factor-α production, 17,611 ± 3,426 vs. 37,486 ± 5,598 pg/mL; p = 0.02). Monocyte function was not suppressed after exposure to MV fractions. RBC supernatants that had been depleted of MVs remained immunosuppressive. Treating RBC supernatants with heat followed by RNase (to degrade protein-bound RNA) prevented RBC supernatant-induced monocyte suppression. CONCLUSION: Our findings implicate soluble mediators of stored RBC-induced monocyte suppression outside of MV fractions and suggest that extracellular protein-bound RNAs (such as microRNA) may play a role in transfusion-related immunomodulation.


Subject(s)
Blood Preservation , Cell-Derived Microparticles/immunology , Culture Media, Conditioned/pharmacology , Erythrocytes/chemistry , Immunosuppression Therapy , Monocytes/immunology , Cells, Cultured , Coculture Techniques , Culture Media/pharmacology , Cytokines/metabolism , Erythrocytes/immunology , Erythrocytes/ultrastructure , Hot Temperature , Humans , In Vitro Techniques , Leukocyte Reduction Procedures , Lipopolysaccharides/pharmacology , RNA/blood , Ribonucleases/pharmacology , Time Factors
17.
J Biol Chem ; 288(6): 3691-5, 2013 Feb 08.
Article in English | MEDLINE | ID: mdl-23269671

ABSTRACT

Burkholderia cenocepacia, the causative agent of cepacia syndrome, primarily affects cystic fibrosis patients, often leading to death. In the lung, epithelial cells serve as the initial barrier to airway infections, yet their responses to B. cenocepacia have not been fully investigated. Here, we examined the molecular responses of human airway epithelial cells to B. cenocepacia infection. Infection led to early signaling events such as activation of Erk, Akt, and NF-κB. Further, TNFα, IL-6, IL-8, and IL-1ß were all significantly induced upon infection, but no IL-1ß was detected in the supernatants. Because caspase-1 is required for IL-1ß processing and release, we examined its expression in airway epithelial cells. Interestingly, little to no caspase-1 was detectable in airway epithelial cells. Transfection of caspase-1 into airway epithelial cells restored their ability to secrete IL-1ß following B. cenocepacia infection, suggesting that a deficiency in caspase-1 is responsible, at least in part, for the attenuated IL-1ß secretion.


Subject(s)
Bronchi/metabolism , Burkholderia Infections/metabolism , Burkholderia cenocepacia , Epithelial Cells/metabolism , Interleukin-1beta/metabolism , Respiratory Mucosa/metabolism , Bronchi/microbiology , Bronchi/pathology , Burkholderia Infections/genetics , Burkholderia Infections/microbiology , Burkholderia Infections/pathology , Caspase 1/biosynthesis , Caspase 1/genetics , Cell Line , Cytokines/biosynthesis , Cytokines/genetics , Epithelial Cells/microbiology , Epithelial Cells/pathology , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Interleukin-1beta/genetics , NF-kappa B/genetics , NF-kappa B/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Respiratory Mucosa/microbiology , Respiratory Mucosa/pathology , Transfection
18.
J Immunol ; 188(7): 3469-77, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22368275

ABSTRACT

Burkholderia cenocepacia is an opportunistic pathogen that causes chronic infection and induces progressive respiratory inflammation in cystic fibrosis patients. Recognition of bacteria by mononuclear cells generally results in the activation of caspase-1 and processing of IL-1ß, a major proinflammatory cytokine. In this study, we report that human pyrin is required to detect intracellular B. cenocepacia leading to IL-1ß processing and release. This inflammatory response involves the host adapter molecule ASC and the bacterial type VI secretion system (T6SS). Human monocytes and THP-1 cells stably expressing either small interfering RNA against pyrin or YFP-pyrin and ASC (YFP-ASC) were infected with B. cenocepacia and analyzed for inflammasome activation. B. cenocepacia efficiently activates the inflammasome and IL-1ß release in monocytes and THP-1. Suppression of pyrin levels in monocytes and THP-1 cells reduced caspase-1 activation and IL-1ß release in response to B. cenocepacia challenge. In contrast, overexpression of pyrin or ASC induced a robust IL-1ß response to B. cenocepacia, which correlated with enhanced host cell death. Inflammasome activation was significantly reduced in cells infected with T6SS-defective mutants of B. cenocepacia, suggesting that the inflammatory reaction is likely induced by an as yet uncharacterized effector(s) of the T6SS. Together, we show for the first time, to our knowledge, that in human mononuclear cells infected with B. cenocepacia, pyrin associates with caspase-1 and ASC forming an inflammasome that upregulates mononuclear cell IL-1ß processing and release.


Subject(s)
Bacterial Secretion Systems/physiology , Burkholderia cenocepacia/immunology , Cytoskeletal Proteins/physiology , Inflammasomes/physiology , Monocytes/microbiology , Apoptosis , Bacterial Secretion Systems/genetics , Burkholderia cenocepacia/genetics , CARD Signaling Adaptor Proteins , Caspase 1/physiology , Cell Line/microbiology , Cytoskeletal Proteins/antagonists & inhibitors , Cytoskeletal Proteins/genetics , Humans , Interleukin-1beta/metabolism , Monocytes/metabolism , Phagocytosis , Pyrin , RNA Interference , RNA, Small Interfering/pharmacology , Recombinant Fusion Proteins/physiology
19.
Mucosal Immunol ; 17(3): 461-475, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38184074

ABSTRACT

Tuberculosis is the leading cause of death for people living with HIV (PLWH). We hypothesized that altered functions of innate immune components in the human alveolar lining fluid of PLWH (HIV-ALF) drive susceptibility to Mycobacterium tuberculosis (M.tb) infection. Our results indicate a significant increase in oxidation of innate proteins and chemokine levels and significantly lower levels and function of complement components and Th1/Th2/Th17 cytokines in HIV-ALF versus control-ALF (non-HIV-infected people). We further found a deficiency of surfactant protein D (SP-D) and reduced binding of SP-D to M.tb that had been exposed to HIV-ALF. Primary human macrophages infected with M.tb exposed to HIV-ALF were significantly less capable of controlling the infection, which was reversed by SP-D replenishment in HIV-ALF. Thus, based on the limited number of participants in this study, our data suggest that PLWH without antiretroviral therapy (ART) have declining host innate defense function in their lung mucosa, thereby favoring M.tb and potentially other pulmonary infections.


Subject(s)
Cytokines , HIV Infections , Immunity, Innate , Mycobacterium tuberculosis , Pulmonary Surfactant-Associated Protein D , Humans , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/physiology , Pulmonary Surfactant-Associated Protein D/metabolism , Pulmonary Surfactant-Associated Protein D/immunology , HIV Infections/immunology , Cytokines/metabolism , Male , Female , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Cells, Cultured , Adult , Tuberculosis, Pulmonary/immunology , Tuberculosis/immunology , Middle Aged , Host-Pathogen Interactions/immunology , Macrophages/immunology , Macrophages/metabolism , Pulmonary Alveoli/immunology , Pulmonary Alveoli/metabolism
20.
J Biol Chem ; 286(5): 3203-8, 2011 Feb 04.
Article in English | MEDLINE | ID: mdl-21097506

ABSTRACT

The ability of Legionella pneumophila to cause pneumonia is determined by its capability to evade the immune system and grow within human monocytes and their derived macrophages. Human monocytes efficiently activate caspase-1 in response to Salmonella but not to L. pneumophila. The molecular mechanism for the lack of inflammasome activation during L. pneumophila infection is unknown. Evaluation of the expression of several inflammasome components in human monocytes during L. pneumophila infection revealed that the expression of the apoptosis-associated speck-like protein (ASC) and the NOD-like receptor NLRC4 are significantly down-regulated in human monocytes. Exogenous expression of ASC maintained the protein level constant during L. pneumophila infection and conveyed caspase-1 activation and restricted the growth of the pathogen. Further depletion of ASC with siRNA was accompanied with improved NF-κB activation and enhanced L. pneumophila growth. Therefore, our data demonstrate that L. pneumophila manipulates ASC levels to evade inflammasome activation and grow in human monocytes. By targeting ASC, L. pneumophila modulates the inflammasome, the apoptosome, and NF-κB pathway simultaneously.


Subject(s)
Cytoskeletal Proteins/physiology , Legionnaires' Disease/immunology , Monocytes/microbiology , Apoptosis , Apoptosis Regulatory Proteins , CARD Signaling Adaptor Proteins , Calcium-Binding Proteins , Caspase 1 , Cytoskeletal Proteins/immunology , Humans , Inflammation , Monocytes/immunology , NF-kappa B/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL