Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Front Immunol ; 8: 522, 2017.
Article in English | MEDLINE | ID: mdl-28533778

ABSTRACT

Pediatric-onset systemic lupus erythematosus arises in humans and mice lacking the endonuclease Dnase1L3. When Dnase1L3 is absent, DNA from circulating apoptotic bodies is not cleared, leading to anti-DNA antibody production. Compared to early anti-DNA and anti-chromatin responses, other autoantibody responses and general immune activation in Dnase1L3-/- mice are greatly delayed. We investigated the possibility that immune activation, specifically inflammasome activation, is regulated by Dnase1L3. Here, we report that Dnase1L3 inhibition blocked both NLR family, pyrin domain containing 3 (NLRP3) and NLRC4 inflammasome-mediated release of high-mobility group box 1 protein and IL-1ß. In contrast to IL-1ß release, Dnase1L3 inhibition only mildly impaired NLRP3-dependent pyroptosis, as measured by propidium iodide uptake or LDH release. Mechanistically, we found that Dnase1L3 was needed to promote apoptosis-associated speck-like protein containing a caspase activation and recruitment domain (ASC) nuclear export and speck formation. Our results demonstrate that Dnase1L3 inhibition separates cytokine secretion from pyroptosis by targeting ASC. These findings suggest that Dnase1L3 is necessary for cytokine secretion following inflammasome activation.

2.
Cancer Immunol Res ; 4(10): 881-892, 2016 10.
Article in English | MEDLINE | ID: mdl-27604597

ABSTRACT

MUC1 is a glycoprotein expressed on the apical surface of ductal epithelial cells. Malignant transformation results in loss of polarization and overexpression of hypoglycosylated MUC1 carrying truncated carbohydrates known as T or Tn tumor antigens. Tumor MUC1 bearing Tn carbohydrates (Tn-MUC1) represent a potential target for immunotherapy. We evaluated the Tn-MUC1 glycopeptide in a human phase I/II clinical trial for safety that followed a preclinical study of different glycosylation forms of MUC1 in rhesus macaques, whose MUC1 is highly homologous to human MUC1. Either unglycosylated rhesus macaque MUC1 peptide (rmMUC1) or Tn-rmMUC1 glycopeptide was mixed with an adjuvant or loaded on autologous dendritic cells (DC), and responses were compared. Unglycosylated rmMUC1 peptide induced negligible humoral or cellular responses compared with the Tn-rmMUC1 glycopeptide. Tn-rmMUC1 loaded on DCs induced the highest anti-rmMUC1 T-cell responses and no clinical toxicity. In the phase I/II clinical study, 17 patients with nonmetastatic castrate-resistant prostate cancer (nmCRPC) were tested with a Tn-MUC1 glycopeptide-DC vaccine. Patients were treated with multiple intradermal and intranodal doses of autologous DCs, which were loaded with the Tn-MUC1 glycopeptide (and KLH as a positive control for immune reactivity). PSA doubling time (PSADT) improved significantly in 11 of 16 evaluable patients (P = 0.037). Immune response analyses detected significant Tn-MUC1-specific CD4+ and/or CD8+ T-cell intracellular cytokine responses in 5 out of 7 patients evaluated. In conclusion, vaccination with Tn-MUC1-loaded DCs in nmCRPC patients appears to be safe, able to induce significant T-cell responses, and have biological activity as measured by the increase in PSADT following vaccination. Cancer Immunol Res; 4(10); 881-92. ©2016 AACR.


Subject(s)
Cancer Vaccines/therapeutic use , Dendritic Cells/transplantation , Mucin-1/immunology , Prostatic Neoplasms, Castration-Resistant/therapy , Aged , Aged, 80 and over , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/adverse effects , Cancer Vaccines/immunology , Feasibility Studies , Humans , Macaca mulatta , Male , Middle Aged , Prostate-Specific Antigen/blood , Prostatic Neoplasms, Castration-Resistant/immunology , Vaccination
3.
J Immunol ; 194(3): 1047-56, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25548234

ABSTRACT

The ability of dendritic cells (DC) to mediate CD4(+) T cell help for cellular immunity is guided by instructive signals received during DC maturation, as well as the resulting pattern of DC responsiveness to the Th signal, CD40L. Furthermore, the professional transfer of antigenic information from migratory DC to lymph node-residing DC is critical for the effective induction of cellular immune responses. In this study we report that, in addition to their enhanced IL-12p70 producing capacity, human DC matured in the presence of inflammatory mediators of type 1 immunity are uniquely programmed to form networks of tunneling nanotube-like structures in response to CD40L-expressing Th cells or rCD40L. This immunologic process of DC reticulation facilitates intercellular trafficking of endosome-associated vesicles and Ag, but also pathogens such HIV-1, and is regulated by the opposing roles of IFN-γ and IL-4. The initiation of DC reticulation represents a novel helper function of CD40L and a superior mechanism of intercellular communication possessed by type 1 polarized DC, as well as a target for exploitation by pathogens to enhance direct cell-to-cell spread.


Subject(s)
CD40 Ligand/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Biological Transport , CD40 Ligand/pharmacology , Cell Communication , Cells, Cultured , Coculture Techniques , Dendritic Cells/drug effects , Dendritic Cells/microbiology , Dendritic Cells/virology , Humans , Inflammation Mediators/metabolism , Lymphocyte Activation/immunology , Signal Transduction , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism
4.
PLoS One ; 9(8): e104210, 2014.
Article in English | MEDLINE | ID: mdl-25117662

ABSTRACT

Regulation of inflammation is necessary to balance sufficient pathogen clearance with excessive tissue damage. Central to regulating inflammation is the switch from a pro-inflammatory pathway to an anti-inflammatory pathway. Macrophages are well-positioned to initiate this switch, and as such are the target of multiple therapeutics. One such potential therapeutic is methylthioadenosine (MTA), which inhibits TNFα production following LPS stimulation. We found that MTA could block TNFα production by multiple TLR ligands. Further, it prevented surface expression of CD69 and CD86 and reduced NF-KB signaling. We then determined that the mechanism of this action by MTA is signaling through adenosine A2 receptors. A2 receptors and TLR receptors synergized to promote an anti-inflammatory phenotype, as MTA enhanced LPS tolerance. In contrast, IL-1ß production and processing was not affected by MTA exposure. Taken together, these data demonstrate that MTA reprograms TLR activation pathways via adenosine receptors to promote resolution of inflammation.


Subject(s)
Deoxyadenosines/pharmacology , Macrophage Activation/immunology , Macrophages/immunology , Macrophages/metabolism , Receptors, Purinergic P1/metabolism , Thionucleosides/pharmacology , Animals , Interleukin-1beta/biosynthesis , Ligands , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Mice , NF-kappa B/metabolism , Purinergic P1 Receptor Agonists/pharmacology , Toll-Like Receptors/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics
5.
J Immunol ; 191(10): 5230-8, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24089192

ABSTRACT

The nucleotide-binding oligomerization domain-like receptor family, pyrin domain-containing 3 (NLRP3) inflammasome drives many inflammatory processes and mediates IL-1 family cytokine release. Inflammasome activators typically damage cells and may release lysosomal and mitochondrial products into the cytosol. Macrophages triggered by the NLRP3 inflammasome activator nigericin show reduced mitochondrial function and decreased cellular ATP. Release of mitochondrial reactive oxygen species (ROS) leads to subsequent lysosomal membrane permeabilization (LMP). NLRP3-deficient macrophages show comparable reduced mitochondrial function and ATP loss, but maintain lysosomal acidity, demonstrating that LMP is NLRP3 dependent. A subset of wild-type macrophages undergo subsequent mitochondrial membrane permeabilization and die. Both LMP and mitochondrial membrane permeabilization are inhibited by potassium, scavenging mitochondrial ROS, or NLRP3 deficiency, but are unaffected by cathepsin B or caspase-1 inhibitors. In contrast, IL-1ß secretion is ablated by potassium, scavenging mitochondrial ROS, and both cathepsin B and caspase-1 inhibition. These results demonstrate interplay between lysosomes and mitochondria that sustain NLRP3 activation and distinguish cell death from IL-1ß release.


Subject(s)
Carrier Proteins/metabolism , Inflammasomes/metabolism , Lysosomes/metabolism , Reactive Oxygen Species/metabolism , Animals , Carrier Proteins/genetics , Caspase 1 , Caspase Inhibitors , Cathepsin B/antagonists & inhibitors , Cells, Cultured , Interleukin-1beta/metabolism , Macrophages , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein , Nigericin , Potassium , Signal Transduction
6.
Toxins (Basel) ; 5(6): 1105-18, 2013 Jun 06.
Article in English | MEDLINE | ID: mdl-23744055

ABSTRACT

Pore-forming toxins are utilized by bacterial and mammalian cells to exert pathogenic effects and induce cell lysis. In addition to rapid plasma membrane repair, macrophages respond to pore-forming toxins through activation of the NLRP3 inflammasome, leading to IL-1ß secretion and pyroptosis. The structural determinants of pore-forming toxins required for NLRP3 activation remain unknown. Here, we demonstrate using streptolysin O (SLO) that pore-formation controls IL-1ß secretion and direct toxicity. An SLO mutant incapable of pore-formation did not promote direct killing, pyroptosis or IL-1ß production. This indicated that pore formation is necessary for inflammasome activation. However, a partially active mutant (SLO N402C) that was less toxic to macrophages than wild-type SLO, even at concentrations that directly lysed an equivalent number of red blood cells, enhanced IL-1ß production but did not alter pyroptosis. This suggests that direct lysis may attenuate immune responses by preventing macrophages from successfully repairing their plasma membrane and elaborating more robust cytokine production. We suggest that mutagenesis of pore-forming toxins represents a strategy to enhance adjuvant activity.


Subject(s)
Inflammasomes/metabolism , Macrophages/drug effects , Streptolysins/genetics , Streptolysins/pharmacology , Animals , Bacterial Proteins/genetics , Bacterial Proteins/pharmacology , CHO Cells , Caspase 1/deficiency , Caspase 1/genetics , Cell Death , Cells, Cultured , Cricetinae , Cricetulus , Interleukin-1beta/metabolism , Lipopolysaccharides , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutagenesis
7.
PLoS One ; 8(4): e61886, 2013.
Article in English | MEDLINE | ID: mdl-23613968

ABSTRACT

Activation of the purinergic receptor P2X7 leads to the cellular permeability of low molecular weight cations. To determine which domains of P2X7 are necessary for this permeability, we exchanged either the C-terminus or portions of the second transmembrane domain (TM2) with those in P2X1 or P2X4. Replacement of the C-terminus of P2X7 with either P2X1 or P2X4 prevented surface expression of the chimeric receptor. Similarly, chimeric P2X7 containing TM2 from P2X1 or P2X4 had reduced surface expression and no permeability to cationic dyes. Exchanging the N-terminal 10 residues or C-terminal 14 residues of the P2X7 TM2 with the corresponding region of P2X1 TM2 partially restored surface expression and limited pore permeability. To further probe TM2 structure, we replaced single residues in P2X7 TM2 with those in P2X1 or P2X4. We identified multiple substitutions that drastically changed pore permeability without altering surface expression. Three substitutions (Q332P, Y336T, and Y343L) individually reduced pore formation as indicated by decreased dye uptake and also reduced membrane blebbing in response to ATP exposure. Three others substitutions, V335T, S342G, and S342A each enhanced dye uptake, membrane blebbing and cell death. Our results demonstrate a critical role for the TM2 domain of P2X7 in receptor function, and provide a structural basis for differences between purinergic receptors.


Subject(s)
Ion Channel Gating , Receptors, Purinergic P2X7/chemistry , Receptors, Purinergic P2X7/metabolism , Adenosine Triphosphate/pharmacology , Amino Acid Substitution/genetics , Animals , Cell Death/drug effects , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Membrane Permeability/drug effects , Cell Survival/drug effects , HEK293 Cells , Humans , Indoles/metabolism , Ion Channel Gating/drug effects , Mice , Mice, Inbred C57BL , Point Mutation/genetics , Protein Structure, Tertiary , Rats , Receptors, Purinergic P2X1/chemistry , Receptors, Purinergic P2X1/metabolism , Receptors, Purinergic P2X4/chemistry , Receptors, Purinergic P2X4/metabolism , Structure-Activity Relationship
8.
J Vis Exp ; (68): e4227, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-23052609

ABSTRACT

Bacterial toxins bind to cholesterol in membranes, forming pores that allow for leakage of cellular contents and influx of materials from the external environment. The cell can either recover from this insult, which requires active membrane repair processes, or else die depending on the amount of toxin exposure and cell type(1). In addition, these toxins induce strong inflammatory responses in infected hosts through activation of immune cells, including macrophages, which produce an array of pro-inflammatory cytokines(2). Many Gram positive bacteria produce cholesterol binding toxins which have been shown to contribute to their virulence through largely uncharacterized mechanisms. Morphologic changes in the plasma membrane of cells exposed to these toxins include their sequestration into cholesterol-enriched surface protrusions, which can be shed into the extracellular space, suggesting an intrinsic cellular defense mechanism(3,4). This process occurs on all cells in the absence of metabolic activity, and can be visualized using EM after chemical fixation(4). In immune cells such as macrophages that mediate inflammation in response to toxin exposure, induced membrane vesicles are suggested to contain cytokines of the IL-1 family and may be responsible both for shedding toxin and disseminating these pro-inflammatory cytokines(5,6,7). A link between IL-1ß release and a specific type of cell death, termed pyroptosis has been suggested, as both are caspase-1 dependent processes(8). To sort out the complexities of this macrophage response, which includes toxin binding, shedding of membrane vesicles, cytokine release, and potentially cell death, we have developed labeling techniques and fluorescence microscopy methods that allow for real time visualization of toxin-cell interactions, including measurements of dysfunction and death (Figure 1). Use of live cell imaging is necessary due to limitations in other techniques. Biochemical approaches cannot resolve effects occurring in individual cells, while flow cytometry does not offer high resolution, real-time visualization of individual cells. The methods described here can be applied to kinetic analysis of responses induced by other stimuli involving complex phenotypic changes in cells.


Subject(s)
Bacterial Toxins/chemistry , Bacterial Toxins/pharmacology , Microscopy, Fluorescence/methods , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/pharmacology , Calcium/metabolism , Dendritic Cells/chemistry , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Erythrocytes/chemistry , Erythrocytes/drug effects , Erythrocytes/metabolism , Fibroblasts/chemistry , Fibroblasts/drug effects , Fibroblasts/metabolism , Hemolysis , Humans , Macrophages/chemistry , Macrophages/drug effects , Macrophages/metabolism , Sheep , Streptolysins/chemistry , Streptolysins/pharmacology
9.
J Immunol ; 189(9): 4621-9, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-23018455

ABSTRACT

Aberrant activation of macrophages in arterial walls by oxidized lipoproteins can lead to atherosclerosis. Oxidized lipoproteins convert macrophages to foam cells through lipid uptake and TLR signaling. To investigate the relative contributions of lipid uptake and TLR signaling in foam cell formation, we established an in vitro assay using liposomes of defined lipid compositions. We found that TLRs signaling through Toll/IL-1R domain-containing adapter inducing IFN-ß promoted foam cell formation by inducing both NF-κB signaling and type I IFN production, whereas TLRs that do not induce IFN, like TLR2, did not enhance foam cell formation. Addition of IFN-α to TLR2 activator promoted robust foam cell formation. TLR signaling further required peroxisome proliferator-activated receptor α, as inhibition of peroxisome proliferator-activated receptor α blocked foam cell formation. We then investigated the ability of endogenous microparticles (MP) to contribute to foam cell formation. We found that lipid-containing MP promoted foam cell formation, which was enhanced by TLR stimulation or IFN-α. These MP also stimulated foam cell formation in a human skin model. However, these MP suppressed TNF-α production and T cell activation, showing that foam cell formation can occur by immunosuppressive MP. Taken together, the data reveal novel signaling requirements for foam cell formation and suggest that uptake of distinct types of MP in the context of activation of multiple distinct TLR can induce foam cell formation.


Subject(s)
Cell Differentiation/immunology , Cell-Derived Microparticles/immunology , Foam Cells/immunology , Foam Cells/metabolism , Macrophages/immunology , Toll-Like Receptors/metabolism , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Cell Line, Tumor , Cell-Derived Microparticles/metabolism , Cell-Derived Microparticles/pathology , Cells, Cultured , Foam Cells/pathology , HeLa Cells , Humans , Ligands , Lipid Metabolism/immunology , Macrophages/metabolism , Macrophages/pathology , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/immunology , Toll-Like Receptors/physiology
10.
J Neuroinflammation ; 9: 84, 2012 May 03.
Article in English | MEDLINE | ID: mdl-22554253

ABSTRACT

BACKGROUND: Immunization against beta-amyloid (Aß) is a promising approach for the treatment of Alzheimer's disease, but the optimal timing for the vaccination remains to be determined. Preventive immunization approaches may be more efficacious and associated with fewer side-effects; however, there is only limited information available from primate models about the effects of preclinical vaccination on brain amyloid composition and the neuroinflammatory milieu. METHODS: Ten non-human primates (NHP) of advanced age (18-26 years) and eight 2-year-old juvenile NHPs were immunized at 0, 2, 6, 10 and 14 weeks with aggregated Aß42 admixed with monophosphoryl lipid A as adjuvant, and monitored for up to 6 months. Anti-Aß antibody levels and immune activation markers were assessed in plasma and cerebrospinal fluid samples before and at several time-points after immunization. Microglial activity was determined by [(11)C]PK11195 PET scans acquired before and after immunization, and by post-mortem immunohistochemical and real-time PCR evaluation. Aß oligomer composition was assessed by immunoblot analysis in the frontal cortex of aged immunized and non-immunized control animals. RESULTS: All juvenile animals developed a strong and sustained serum anti-Aß IgG antibody response, whereas only 80 % of aged animals developed detectable antibodies. The immune response in aged monkeys was more delayed and significantly weaker, and was also more variable between animals. Pre- and post-immunization [(11)C]PK11195 PET scans showed no evidence of vaccine-related microglial activation. Post-mortem brain tissue analysis indicated a low overall amyloid burden, but revealed a significant shift in oligomer size with an increase in the dimer:pentamer ratio in aged immunized animals compared with non-immunized controls (P < 0.01). No differences were seen in microglial density or expression of classical and alternative microglial activation markers between immunized and control animals. CONCLUSIONS: Our results indicate that preventive Aß immunization is a safe therapeutic approach lacking adverse CNS immune system activation or other serious side-effects in both aged and juvenile NHP cohorts. A significant shift in the composition of soluble oligomers towards smaller species might facilitate removal of toxic Aß species from the brain.


Subject(s)
Aging/immunology , Alzheimer Vaccines/administration & dosage , Alzheimer Vaccines/immunology , Amyloid beta-Peptides/administration & dosage , Amyloid beta-Peptides/immunology , Brain/immunology , Immunization/methods , Peptide Fragments/administration & dosage , Peptide Fragments/immunology , Age Factors , Aging/metabolism , Alzheimer Vaccines/therapeutic use , Amyloid beta-Peptides/metabolism , Animals , Brain/metabolism , Female , Macaca fascicularis , Macaca mulatta , Macaca nemestrina , Male , Peptide Fragments/metabolism
11.
Angew Chem Int Ed Engl ; 51(20): 4838-42, 2012 May 14.
Article in English | MEDLINE | ID: mdl-22461279

ABSTRACT

Traffic cam: a tandem dye prepared from a FRET acceptor and a fluorogenic donor functions as a cell surface ratiometric pH indicator, which upon internalization serves to follow protein trafficking during endocytosis. This sensor was used to analyze agonist-dependent internalization of ß(2)-adrenergic receptors. It was also used as a surrogate antigen to reveal direct surface-to-endosome antigen transfer between dendritic cells (not shown).


Subject(s)
Biosensing Techniques/methods , Endocytosis/physiology , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Animals , Antigen Presentation , Dendritic Cells/chemistry , Dendritic Cells/immunology , Dendritic Cells/metabolism , Fluorescence Resonance Energy Transfer , Fluorescent Dyes/chemistry , Hydrogen-Ion Concentration , Mice , NIH 3T3 Cells , Protein Transport
12.
PLoS One ; 6(7): e22401, 2011.
Article in English | MEDLINE | ID: mdl-21789257

ABSTRACT

Viral and bacterial infections of the lower respiratory tract are major causes of morbidity and mortality worldwide. Alveolar macrophages line the alveolar spaces and are the first cells of the immune system to respond to invading pathogens. To determine the similarities and differences between the responses of mice and macaques to invading pathogens we profiled alveolar macrophages from these species following infection with two viral (PR8 and Fuj/02 influenza A) and two bacterial (Mycobacterium tuberculosis and Francisella tularensis Schu S4) pathogens. Cells were collected at 6 time points following each infection and expression profiles were compared across and between species. Our analyses identified a core set of genes, activated in both species and across all pathogens that were predominantly part of the interferon response pathway. In addition, we identified similarities across species in the way innate immune cells respond to lethal versus non-lethal pathogens. On the other hand we also found several species and pathogen specific response patterns. These results provide new insights into mechanisms by which the innate immune system responds to, and interacts with, invading pathogens.


Subject(s)
Bacteria/immunology , Host-Pathogen Interactions/immunology , Immunity, Innate/genetics , Immunity, Innate/immunology , Macaca/microbiology , Macaca/virology , Viruses/immunology , Animals , Francisella tularensis/immunology , Gene Expression Profiling , Gene Expression Regulation , Host-Pathogen Interactions/genetics , Influenza A virus/immunology , Interferon Regulatory Factor-7/genetics , Interferon Regulatory Factor-7/metabolism , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/microbiology , Macrophages, Alveolar/virology , Mice , Mycobacterium tuberculosis/immunology , Oligonucleotide Array Sequence Analysis , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/virology , Signal Transduction/genetics , Species Specificity , Tuberculosis/genetics , Tuberculosis/microbiology , Tularemia/genetics , Tularemia/microbiology , Up-Regulation
13.
Immunol Res ; 50(2-3): 118-23, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21717083

ABSTRACT

Toxins secreted by bacteria can impact the host in a number of different ways. In some infections, toxins play a crucial and central role in pathogenesis (i.e., anthrax), while in other bacterial infections, the role of toxins is less understood. The cholesterol-dependent cytolysins (CDCs), of which streptolysin O is a prototype, are a class of pore-forming toxins produced by many gram-positive bacteria and have only been studied in a few experimental infection models. Our laboratory has demonstrated that CDCs have effects on macrophages that are both pro- and anti-inflammatory. Here, we review evidence that CDCs promote inflammation by driving secretion of IL-1ß and HMGB-1 from macrophages in a NLRP3-dependent manner, while also causing shedding of membrane microvesicles from cells that can interact with macrophages and inhibit TNF-α release. CDCs thus impact macrophage function in ways that may be both beneficial and detrimental to the host.


Subject(s)
Bacterial Toxins/metabolism , Macrophages/immunology , Macrophages/metabolism , Animals , Bacterial Toxins/pharmacology , Cholesterol/metabolism , Cytotoxins/immunology , Cytotoxins/metabolism , Humans , Inflammation/immunology , Inflammation/metabolism , Macrophages/drug effects , Myeloid Cells/immunology , Myeloid Cells/metabolism , Pore Forming Cytotoxic Proteins/metabolism , Tetanus Toxin/metabolism , Tetanus Toxin/pharmacology
14.
Am J Pathol ; 178(6): 2752-9, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21641397

ABSTRACT

Extracellular superoxide dismutase (EC-SOD) is abundant in the lung and limits inflammation and injury in response to many pulmonary insults. To test the hypothesis that EC-SOD has an important role in bacterial infections, wild-type and EC-SOD knockout (KO) mice were infected with Escherichia coli to induce pneumonia. Although mice in the EC-SOD KO group demonstrated greater pulmonary inflammation than did wild-type mice, there was less clearance of bacteria from their lungs after infection. Macrophages and neutrophils express EC-SOD; however, its function and subcellular localization in these inflammatory cells is unclear. In the present study, immunogold electron microscopy revealed EC-SOD in membrane-bound vesicles of phagocytes. These findings suggest that inflammatory cell EC-SOD may have a role in antibacterial defense. To test this hypothesis, phagocytes from wild-type and EC-SOD KO mice were evaluated. Although macrophages lacking EC-SOD produced more reactive oxygen species than did cells expressing EC-SOD after stimulation, they demonstrated significantly impaired phagocytosis and killing of bacteria. Overall, this suggests that EC-SOD facilitates clearance of bacteria and limits inflammation in response to infection by promoting bacterial phagocytosis.


Subject(s)
Escherichia coli/cytology , Extracellular Space/enzymology , Macrophages/cytology , Macrophages/enzymology , Microbial Viability , Phagocytosis , Superoxide Dismutase/metabolism , Animals , Humans , Inflammation/microbiology , Inflammation/pathology , Intracellular Space/metabolism , Lung/microbiology , Lung/pathology , Macrophages/ultrastructure , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidants/metabolism , Pneumonia/microbiology , Pneumonia/pathology , Superoxide Dismutase/ultrastructure
15.
J Cell Sci ; 124(Pt 14): 2414-23, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21693578

ABSTRACT

Cells survive exposure to bacterial pore-forming toxins, such as streptolysin O (SLO), through mechanisms that remain unclear. Previous studies have suggested that these toxins are cleared by endocytosis. However, the experiments reported here failed to reveal any evidence for endocytosis of SLO, nor did they reveal any signs of damage to endosomal membranes predicted from such endocytosis. Instead, we illustrate that SLO induces a characteristic form of plasma membrane blebbing that allows cells to shed SLO by the process known as ectocytosis. Specifically, 'deep-etch' electron microscopy of cells exposed to SLO illustrates that the toxin is rapidly sequestered into domains in the plasmalemma greatly enriched in SLO pores, and these domains bleb outwards and bud from the cell surface into the medium. Such ectocytosis is even observed in cells that have been chemically fixed before exposure to SLO, suggesting that it is caused by a direct physical action of the toxin on the cell membrane, rather than by an active cellular reaction. We conclude, therefore, that ectocytosis is an important means for SLO clearance and hypothesize that this is a primary method by which cells defend themselves generally against pore-forming toxins.


Subject(s)
Streptolysins/metabolism , Animals , Bacterial Proteins/metabolism , CHO Cells , Cell Membrane/metabolism , Cell Membrane Structures/metabolism , Cell Survival/physiology , Cricetinae , Cricetulus , Endocytosis , Humans
16.
J Immunol ; 185(6): 3740-9, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20709956

ABSTRACT

ATP-mediated activation of the purinergic receptor P2X(7) elicits morphological changes and proinflammatory responses in macrophages. These changes include rapid shedding of microvesicles (MV) and the nonconventional secretion of cytokines, such as IL-1beta and IL-18 following priming. In this study, we demonstrate the activation potential of P2X(7)-induced MV isolated from nonprimed murine macrophages. Cotreatment of nonprimed macrophages with ATP and calcium ionophore induced a rapid release of MV that were predominantly 0.5-1 microm in size. Exposure of primary murine bone marrow-derived macrophages to these MV resulted in costimulatory receptor upregulation and TNF-alpha secretion. Cell homogenates or supernatants cleared of MV did not activate macrophages. MV-mediated activation was p38 MAPK and NF-kappaB dependent, and partially dependent on TLR4 activity, but was high-mobility group box 1 independent. Biochemical fractionation of the MV demonstrated that the phospholipid fraction, not the protein fraction, mediated macrophage activation through a TLR4-dependent process. P2X(7) activation is known to induce calcium-independent phospholipase A(2), calcium-dependent phospholipase A(2), and phospholipase D activities, but inhibition of these enzymes did not inhibit MV generation or shedding. However, blocking phospholipase D activity resulted in release of MV incapable of activating recipient macrophages. These data demonstrate a novel mechanism of macrophage activation resulting from exposure to MV from nonprimed macrophages, and identifies phospholipids in these MV as the biologically active component. We suggest that phospholipids delivered by MV may be mediators of sterile inflammation in a number of diseases.


Subject(s)
Macrophage Activation/immunology , Macrophages/immunology , Myeloid Cells/immunology , Phospholipids/physiology , Receptors, Purinergic P2X7/physiology , Secretory Vesicles/immunology , Toll-Like Receptor 4/physiology , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cell Line , HMGB1 Protein/physiology , Humans , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/metabolism , NF-kappa B/physiology , Secretory Vesicles/metabolism , Signal Transduction/immunology , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/genetics , p38 Mitogen-Activated Protein Kinases/physiology
17.
Purinergic Signal ; 6(1): 31-45, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19838818

ABSTRACT

The P2X(7) receptor exhibits significant allelic polymorphism in humans, with both loss and gain of function variants potentially impacting on a variety of infectious and inflammatory disorders. At least five loss-of-function polymorphisms (G150R, R307Q, T357S, E496A, and I568N) and two gain-of-function polymorphisms (H155Y and Q460R) have been identified and characterized to date. In this study, we used RT-PCR cloning to isolate and characterize P2X(7) cDNA clones from human PBMCs and THP-1 cells. A previously unreported variant with substitutions of V80M and A166G was identified. When expressed in HEK293 cells, this variant exhibited heightened sensitivity to the P2X(7) agonist (BzATP) relative to the most frequent allele, as shown by pore formation measured by fluorescent dye uptake into cells. Mutational analyses showed that A166G alteration was critical for the gain-of-function change, while V80M was not. Full-length variants with multiple previously identified nonsynonymous SNPs (H155Y, H270R, A348T, and E496A) were also identified. Distinct functional phenotypes of the P2X(7) variants or mutants constructed with multiple polymorphisms were observed. Gain-of-function variations (A166G or H155Y) could not rescue the loss-of-function E496A polymorphism. Synergistic effects of the gain-of-function variations were also observed. We also identified the A348T alteration as a weak gain-of-function variant. Thus, these results identify the new gain-of-function variant A166G and demonstrate that multiple-gene polymorphisms contribute to functional phenotypes of the human P2X(7) receptor. Furthermore, the results demonstrate that the C-terminal of the cysteine-rich domain 1 of P2X(7) is critical for regulation of P2X(7)-mediated pore formation.

18.
Immunol Lett ; 2009 Oct 04.
Article in English | MEDLINE | ID: mdl-19808050

ABSTRACT

This article has been withdrawn at the request of the authors and Guest Editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at http://www.elsevier.com/locate/withdrawalpolicy.

19.
Immunol Rev ; 231(1): 278-88, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19754904

ABSTRACT

Ca2+-driven responses in dendritic cells (DCs) are less well characterized than in lymphocytes. When DCs undergo a sequence of activation/maturation events, typically beginning with exposure to pathogens in the periphery, Ca2+ entry into the cytosol from stores in the endoplasmic reticulum or from outside the cell can occur at various steps and participate in intracellular signaling. However, not all cellular processes identified in these cells are Ca2+ dependent. While immigration of precursor DCs into the peripheral tissues as well as emigration to secondary lymphoid sites following microbial challenge depend on processes that involve Ca2+, other processes such as DC maturation in response to Toll-like receptor agonist stimulation appear not to. Certain microbial stimuli and host-derived chemokines induce Ca2+ entry that is important for the induced responses. In this article, we review the current state of our understanding of the role of Ca2+ in DC biology and argue that homeostatic control of Ca2+ levels in these cells is critical for maintaining their proper function. We also consider evidence for intercellular transmission of Ca2+ signals between DCs that are physically linked by thin membranous extensions termed tunneling nanotubules.


Subject(s)
Calcium/metabolism , Dendritic Cells/metabolism , Animals , Antigens, Bacterial/immunology , Bacteria/immunology , Calcium Channels/metabolism , Cell Communication , Dendritic Cells/immunology , Humans
20.
J Leukoc Biol ; 86(5): 1227-38, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19675207

ABSTRACT

CDC are exotoxins secreted by many Gram-positive bacteria that bind cholesterol and oligomerize to form pores in eukaryotic cell membranes. We demonstrate that CDC TLO induces caspase-1 cleavage and the rapid release of IL-1beta from LPS-primed murine BMDM. IL-1beta secretion depends on functional toxin pore formation, as free cholesterol, which prevents TLO binding to cell membranes, blocks the cytokine release. Secretion of the mature forms of IL-1beta and caspase-1 occurs only at lower TLO doses, whereas at a higher concentration, cells release the biologically inactive proforms. IL-1beta release at a low TLO dose requires potassium efflux, calcium influx, and the activities of calcium-independent PLA(2), caspase-1, and cathepsin B. Additionally, mature IL-1beta release induced by a low TLO dose is dependent on the NLRP3 inflammasome, and pro-IL-1beta release induced by a high TLO dose occurs independently of NLRP3. These results further elucidate a mechanism of CDC-induced IL-1beta release and suggest a novel, immune evasion strategy in which IL-1beta-containing macrophages might release primarily inactive cytokine following exposure to high doses of these toxins.


Subject(s)
Carrier Proteins/physiology , Cathepsin B/physiology , Cholesterol/pharmacology , Cytotoxins/pharmacology , Exotoxins/toxicity , Interleukin-1beta/metabolism , Macrophages/metabolism , Animals , Cytotoxins/toxicity , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gram-Positive Bacteria/physiology , Inflammation/blood , Inflammation/physiopathology , Lipopolysaccharides/pharmacology , Macrophages/cytology , Macrophages/drug effects , Mice , NLR Family, Pyrin Domain-Containing 3 Protein , Pepstatins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...