Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
J Biol Chem ; 287(39): 32791-9, 2012 Sep 21.
Article in English | MEDLINE | ID: mdl-22843689

ABSTRACT

The interleukin (IL)-1ß-processing inflammasome has recently been identified as a target for pathogenic evasion of the inflammatory response by a number of bacteria and viruses. We postulated that the periodontal pathogen, Porphyromonas gingivalis may suppress the inflammasome as a mechanism for its low immunogenicity and pathogenic synergy with other, more highly immunogenic periodontal bacteria. Our results show that P. gingivalis lacks signaling capability for the activation of the inflammasome in mouse macrophages. Furthermore, P. gingivalis can suppress inflammasome activation by another periodontal bacterium, Fusobacterium nucleatum. This repression affects IL-1ß processing, as well as other inflammasome-mediated processes, including IL-18 processing and cell death, in both human and mouse macrophages. F. nucleatum activates IL-1ß processing through the Nlrp3 inflammasome; however, P. gingivalis repression is not mediated through reduced levels of inflammasome components. P. gingivalis can repress Nlrp3 inflammasome activation by Escherichia coli, and by danger-associated molecular patterns and pattern-associated molecular patterns that mediate activation through endocytosis. However, P. gingivalis does not suppress Nlrp3 inflammasome activation by ATP or nigericin. This suggests that P. gingivalis may preferentially suppress endocytic pathways toward inflammasome activation. To directly test whether P. gingivalis infection affects endocytosis, we assessed the uptake of fluorescent particles in the presence or absence of P. gingivalis. Our results show that P. gingivalis limits both the number of cells taking up beads and the number of beads taken up for bead-positive cells. These results provide a novel mechanism of pathogen-mediated inflammasome inhibition through the suppression of endocytosis.


Subject(s)
Bacteroidaceae Infections/immunology , Endocytosis/immunology , Inflammasomes/immunology , Macrophage Activation/immunology , Macrophages/immunology , Porphyromonas gingivalis/immunology , Animals , Carrier Proteins/immunology , Cells, Cultured , Coculture Techniques , Escherichia coli/immunology , Fusobacterium/immunology , Humans , Macrophages/microbiology , Macrophages/pathology , Mice , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein
2.
Nat Immunol ; 12(5): 408-15, 2011 May.
Article in English | MEDLINE | ID: mdl-21478880

ABSTRACT

High-fat diet (HFD) and inflammation are key contributors to insulin resistance and type 2 diabetes (T2D). Interleukin (IL)-1ß plays a role in insulin resistance, yet how IL-1ß is induced by the fatty acids in an HFD, and how this alters insulin signaling, is unclear. We show that the saturated fatty acid palmitate, but not unsaturated oleate, induces the activation of the NLRP3-ASC inflammasome, causing caspase-1, IL-1ß and IL-18 production. This pathway involves mitochondrial reactive oxygen species and the AMP-activated protein kinase and unc-51-like kinase-1 (ULK1) autophagy signaling cascade. Inflammasome activation in hematopoietic cells impairs insulin signaling in several target tissues to reduce glucose tolerance and insulin sensitivity. Furthermore, IL-1ß affects insulin sensitivity through tumor necrosis factor-independent and dependent pathways. These findings provide insights into the association of inflammation, diet and T2D.


Subject(s)
Carrier Proteins/immunology , Dietary Fats/immunology , Inflammasomes/immunology , Insulin Resistance/immunology , Palmitic Acid/immunology , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Animals , Autophagy/immunology , Caspase 1/immunology , Enzyme Activation , Enzyme Inhibitors/pharmacology , Flow Cytometry , Interleukin-1beta/immunology , Macrophages , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , NLR Family, Pyrin Domain-Containing 3 Protein , Oligopeptides/pharmacology , Reactive Oxygen Species/immunology , Ribonucleotides/pharmacology , Signal Transduction
3.
J Biol Chem ; 286(22): 19605-16, 2011 Jun 03.
Article in English | MEDLINE | ID: mdl-21487011

ABSTRACT

ASC/PYCARD is a common adaptor for a diverse set of inflammasomes that activate caspase-1, most prominently the NLR-based inflammasome. Mounting evidence indicates that ASC and these NLRs also elicit non-overlapping functions, but the molecular basis for this difference is unclear. To address this, we performed microarray and network analysis of ASC shRNA knockdown cells. In pathogen-infected cells, an ASC-dependent interactome is centered on the mitogen-activated protein kinase (MAPK) ERK and on multiple chemokines. ASC did not affect the expression of MAPK but affected its phosphorylation by pathogens and Toll-like receptor agonists via suppression of the dual-specificity phosphatase, DUSP10/MKP5. Chemokine induction, DUSP function, and MAPK phosphorylation were independent of caspase-1 and IL-1ß. MAPK activation by pathogen was abrogated in Asc(-/-) but not Nlrp3(-/-), Nlrc4(-/-), or Casp1(-/-) macrophages. These results demonstrate a function for ASC that is distinct from the inflammasome in modulating MAPK activity and chemokine expression and further identify DUSP10 as a novel ASC target.


Subject(s)
Chemokines/biosynthesis , Cytoskeletal Proteins/metabolism , Dual-Specificity Phosphatases/metabolism , Inflammasomes/metabolism , Macrophages/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinase Phosphatases/metabolism , Animals , Apoptosis Regulatory Proteins , CARD Signaling Adaptor Proteins , Cell Line , Chemokines/genetics , Cytoskeletal Proteins/genetics , Dual-Specificity Phosphatases/genetics , Enzyme Activation/physiology , Gene Knockdown Techniques , Humans , Inflammasomes/genetics , Macrophages/cytology , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinase Phosphatases/genetics
4.
J Immunol ; 185(9): 5476-85, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20921527

ABSTRACT

Francisella tularensis is a facultative intracellular pathogen and potential biothreat agent. Evasion of the immune response contributes to the extraordinary virulence of this organism although the mechanism is unclear. Whereas wild-type strains induced low levels of cytokines, an F. tularensis ripA deletion mutant (LVSΔripA) provoked significant release of IL-1ß, IL-18, and TNF-α by resting macrophages. IL-1ß and IL-18 secretion was dependent on inflammasome components pyrin-caspase recruitment domain/apoptotic speck-containing protein with a caspase recruitment domain and caspase-1, and the TLR/IL-1R signaling molecule MyD88 was required for inflammatory cytokine synthesis. Complementation of LVSΔripA with a plasmid encoding ripA restored immune evasion. Similar findings were observed in a human monocytic line. The presence of ripA nearly eliminated activation of MAPKs including ERK1/2, JNK, and p38, and pharmacologic inhibitors of these three MAPKs reduced cytokine induction by LVSΔripA. Animals infected with LVSΔripA mounted a stronger IL-1ß and TNF-α response than that of mice infected with wild-type live vaccine strain. This analysis revealed novel immune evasive mechanisms of F. tularensis.


Subject(s)
Francisella tularensis/pathogenicity , Genes, Bacterial/immunology , Inflammation/genetics , Macrophages/immunology , Mitogen-Activated Protein Kinases/genetics , Signal Transduction/genetics , Animals , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Bacterial Proteins/metabolism , Blotting, Western , Cytokines/biosynthesis , Enzyme-Linked Immunosorbent Assay , Female , Francisella tularensis/genetics , Francisella tularensis/immunology , Genes, Bacterial/genetics , Humans , Immune Evasion/genetics , Immune Evasion/immunology , Inflammation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinases/immunology , Signal Transduction/immunology , Tularemia/genetics , Tularemia/immunology
5.
Methods Mol Biol ; 629: 141-58, 2010.
Article in English | MEDLINE | ID: mdl-20387148

ABSTRACT

Shortly after the cellular mechanism of RNA interference (RNAi) was first described, scientists began using this powerful technique to study gene function. This included designing better methods for the successful delivery of small interfering RNAs (siRNAs) and short hairpin RNAs (shRNAs) into mammalian cells. While the simplest method for RNAi is the cytosolic delivery of siRNA oligonucleotides, this technique is limited to cells capable of transfection and is primarily utilized during transient in vitro studies. The introduction of shRNA into mammalian cells through infection with viral vectors allows for stable integration of shRNA and long-term knockdown of the targeted gene; however, several challenges exist with the implementation of this technology. Here we describe some well-tested protocols which should increase the chances of successful design, delivery, and assessment of gene knockdown by shRNA. We provide suggestions for designing shRNA targets and controls, a protocol for sequencing through the secondary structure of the shRNA hairpin structure, and protocols for packaging and delivery of shRNA lentiviral particles. Using real-time PCR and functional assays we demonstrate the successful knockdown of ASC, an inflammatory adaptor molecule. These studies demonstrate the practicality of including two shRNAs with different efficacies of knockdown to provide an additional level of control and to verify dose dependency of functional effects. Along with the methods described here, as new techniques and algorithms are designed in the future, shRNA is likely to include further promising application and continue to be a critical component of gene discovery.


Subject(s)
Gene Knockdown Techniques/methods , RNA, Small Interfering/chemical synthesis , RNA, Small Interfering/metabolism , Adaptor Proteins, Signal Transducing/genetics , Cell Adhesion , Cell Line , Enzyme-Linked Immunosorbent Assay , Humans , Interleukin-1beta/metabolism , Lentivirus/physiology , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, RNA , Transduction, Genetic , Virus Assembly
6.
J Immunol ; 183(3): 2008-15, 2009 Aug 01.
Article in English | MEDLINE | ID: mdl-19587006

ABSTRACT

Bacterial infection elicits a range of beneficial as well as detrimental host inflammatory responses. Key among these responses are macrophage/monocyte necrosis, release of the proinflammatory factor high-mobility group box 1 protein (HMGB1), and induction of the cytokine IL-1. Although the control of IL-1beta has been well studied, processes that control macrophage cell death and HMGB1 release in animals are poorly understood. This study uses Klebsiella pneumonia as a model organism because it elicits all three responses in vivo. The regulation of these responses is studied in the context of the inflammasome components NLRP3 and ASC, which are important for caspase-1 activation and IL-1beta release. Using a pulmonary infection model that reflects human infection, we show that K. pneumonia-induced mouse macrophage necrosis, HMGB1, and IL-1beta release are dependent on NLRP3 and ASC. K. pneumoniae infection of mice lacking Nlrp3 results in decreased lung inflammation and reduced survival relative to control, indicating the overall protective role of this gene. Macrophage/monocyte necrosis and HMGB1 release are controlled independently of caspase-1, suggesting that the former two responses are separable from inflammasome-associated functions. These results provide critical in vivo validation that the physiologic role of NLRP3 and ASC is not limited to inflammasome formation.


Subject(s)
Carrier Proteins/physiology , Caspase 1/metabolism , Cytoskeletal Proteins/physiology , HMGB1 Protein/metabolism , Pneumonia/metabolism , Animals , Apoptosis Regulatory Proteins , CARD Signaling Adaptor Proteins , Disease Models, Animal , Inflammation/metabolism , Inflammation/microbiology , Interleukin-1beta/metabolism , Klebsiella , Macrophages/microbiology , Macrophages/pathology , Mice , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Necrosis , Pneumonia/microbiology , Pneumonia/pathology
7.
J Immunol ; 182(10): 6460-9, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19414800

ABSTRACT

Neisseria gonorrhoeae is a common sexually transmitted pathogen that significantly impacts female fertility, neonatal health, and transmission of HIV worldwide. N. gonorrhoeae usually causes localized inflammation of the urethra and cervix by inducing production of IL-1beta and other inflammatory cytokines. Several NLR (nucleotide-binding domain, leucine-rich repeat) proteins are implicated in the formation of pro-IL-1beta-processing complexes called inflammasomes in response to pathogens. We demonstrate that NLRP3 (cryopyrin, NALP3) is the primary NLR required for IL-1beta/IL-18 secretion in response to N. gonorrhoeae in monocytes. We also show that N. gonorrhoeae infection promotes NLRP3-dependent monocytic cell death via pyronecrosis, a recently described pathway with morphological features of necrosis, including release of the strong inflammatory mediator HMBG1. Additionally, N. gonorrhoeae activates the cysteine protease cathepsin B as measured by the breakdown of a cathepsin B substrate. Inhibition of cathepsin B shows that this protease is an apical controlling step in the downstream activities of NLRP3 including IL-1beta production, pyronecrosis, and HMGB1 release. Nonpathogenic Neisseria strains (Neisseria cinerea and Neisseria flavescens) do not activate NLRP3 as robustly as N. gonorrhoeae. Conditioned medium from N. gonorrhoeae contains factors capable of initiating the NLRP3-mediated signaling events. Isolated N. gonorrhoeae lipooligosaccharide, a known virulence factor from this bacterium that is elaborated from the bacterium in the form of outer membrane blebs, activates both NLRP3-induced IL-1beta secretion and pyronecrosis. Our findings indicate that activation of NLRP3-mediated inflammatory response pathways is an important venue associated with host response and pathogenesis of N. gonorrhoeae.


Subject(s)
Carrier Proteins/immunology , Cathepsin B/immunology , Cytoskeletal Proteins/immunology , Inflammation/immunology , Neisseria gonorrhoeae/immunology , Signal Transduction/immunology , Animals , Apoptosis Regulatory Proteins , Blotting, Western , CARD Signaling Adaptor Proteins , Carrier Proteins/metabolism , Cathepsin B/metabolism , Cytokines/immunology , Cytokines/metabolism , Cytoskeletal Proteins/metabolism , Enzyme Activation/immunology , Enzyme-Linked Immunosorbent Assay , Female , HMGB1 Protein/immunology , HMGB1 Protein/metabolism , Humans , Inflammation/metabolism , Interleukin-18/immunology , Interleukin-18/metabolism , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Male , Mice , Mice, Knockout , Monocytes/immunology , NLR Family, Pyrin Domain-Containing 3 Protein , Necrosis/immunology , RNA, Small Interfering
8.
J Immunol ; 182(4): 2395-404, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-19201894

ABSTRACT

Periodontal disease is a chronic inflammatory disorder that leads to the destruction of tooth-supporting tissue and affects 10-20 million people in the U.S. alone. The oral pathogen Porphyromonas gingivalis causes inflammatory host response leading to periodontal and other secondary inflammatory diseases. To identify molecular components that control host response to P. gingivalis in humans, roles for the NLR (NBD-LRR) protein, NLRP3 (cryopyrin, NALP3), and its adaptor apoptotic speck protein containing a C-terminal caspase recruitment domain (ASC) were studied. P. gingivalis strain A7436 induces cell death in THP1 monocytic cells and in human primary peripheral blood macrophages. This process is ASC and NLRP3 dependent and can be replicated by P. gingivalis LPS and Escherichia coli. P. gingivalis-induced cell death is caspase and IL-1 independent and exhibits morphological features consistent with necrosis including loss of membrane integrity and release of cellular content. Intriguingly, P. gingivalis-induced cell death is accompanied by the formation of ASC aggregation specks, a process not previously described during microbial infection. ASC specks are observed in P. gingivalis-infected primary human mononuclear cells and are dependent on NLRP3. This work shows that P. gingivalis causes ASC- and NLRP3-dependent necrosis, accompanied by ASC speck formation.


Subject(s)
Bacteroidaceae Infections/metabolism , Carrier Proteins/metabolism , Cytoskeletal Proteins/metabolism , Macrophages/microbiology , Monocytes/microbiology , Necrosis/metabolism , Bacteroidaceae Infections/immunology , Bacteroidaceae Infections/pathology , Blotting, Western , CARD Signaling Adaptor Proteins , Carrier Proteins/immunology , Cytoskeletal Proteins/immunology , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Humans , Macrophages/immunology , Macrophages/metabolism , Microscopy, Electron, Transmission , Monocytes/immunology , Monocytes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein , Necrosis/immunology , Necrosis/microbiology , Porphyromonas gingivalis , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...