Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
Infect Immun ; 85(11)2017 11.
Article in English | MEDLINE | ID: mdl-28808162

ABSTRACT

Activation of the innate immune receptor NLRP1B leads to the formation of an inflammasome, which induces autoproteolytic processing of pro-caspase-1, and ultimately to the release of inflammatory cytokines and to the execution of pyroptosis. One of the signals to which NLRP1B responds is metabolic stress that occurs in cells deprived of glucose or treated with metabolic inhibitors. NLRP1B might therefore sense microbial infection, as intracellular pathogens such as Listeria monocytogenes and Shigella flexneri cause metabolic stress as a result of nutrient scavenging and host cell damage. Here we addressed whether these pathogens activate the NLRP1B inflammasome. We found that Listeria infection activated the NLRP1B inflammasome in a reconstituted fibroblast model. Activation of NLRP1B by Listeria was diminished in an NLRP1B mutant shown previously to be defective at detecting energy stress and was dependent on the expression of listeriolysin O (LLO), a protein required for vacuolar escape. Infections of either Listeria or Shigella activated NLRP1B in the RAW264.7 murine macrophage line, which expresses endogenous NLRP1B. We conclude that NLRP1B senses cellular infection by distinct invasive pathogens.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Bacterial Toxins/genetics , Heat-Shock Proteins/genetics , Hemolysin Proteins/genetics , Inflammasomes/genetics , Listeria monocytogenes/genetics , Shigella flexneri/genetics , Animals , Apoptosis Regulatory Proteins/deficiency , Apoptosis Regulatory Proteins/immunology , Bacterial Toxins/metabolism , Cell Line , Cell Line, Tumor , Fibroblasts/immunology , Fibroblasts/microbiology , Gene Expression Regulation , Heat-Shock Proteins/metabolism , Hemolysin Proteins/metabolism , Host-Pathogen Interactions , Humans , Inflammasomes/immunology , Listeria monocytogenes/growth & development , Listeria monocytogenes/metabolism , Macrophages/immunology , Macrophages/microbiology , Mice , Mutation , Shigella flexneri/growth & development , Shigella flexneri/metabolism , Signal Transduction
2.
J Immunol ; 194(12): 5663-73, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25964492

ABSTRACT

Type 1 diabetes in the NOD mouse model has been linked to >30 insulin-dependent diabetes (Idd) susceptibility loci. Idd4 on chromosome 11 consists of two subloci, Idd4.1 and Idd4.2. Using congenic analysis of alleles in NOD and NOD-resistant (NOR) mice, we previously defined Idd4.1 as an interval containing >50 genes that controlled expression of genes in the type 1 IFN pathway. In this study, we report refined mapping of Idd4.1 to a 1.1-Mb chromosomal region and provide genomic sequence analysis and mechanistic evidence supporting its role in innate immune regulation of islet-directed autoimmunity. Genetic variation at Idd4.1 was mediated by radiation-sensitive hematopoietic cells, and type 1 diabetes protection conferred by the NOR allele was abrogated in mice treated with exogenous type 1 IFN-ß. Next generation sequence analysis of the full Idd4.1 genomic interval in NOD and NOR strains supported Nlrp1b as a strong candidate gene for Idd4.1. Nlrp1b belongs to the Nod-like receptor (NLR) gene family and contributes to inflammasome assembly, caspase-1 recruitment, and release of IL-1ß. The Nlrp1b of NOR was expressed as an alternative spliced isoform that skips exon 9, resulting in a premature stop codon predicted to encode a truncated protein. Functional analysis of the truncated NOR Nlrp1b protein demonstrated that it was unable to recruit caspase-1 and process IL-1ß. Our data suggest that Idd4.1-dependent protection from islet autoimmunity is mediated by differences in type 1 IFN- and IL-1ß-dependent immune responses resulting from genetic variation in Nlrp1b.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Diabetes Mellitus, Type 1/genetics , Genetic Predisposition to Disease , Inflammasomes/genetics , Quantitative Trait Loci , Alleles , Alternative Splicing , Animals , Apoptosis Regulatory Proteins/chemistry , Base Sequence , Chromosome Mapping , Diabetes Mellitus, Type 1/immunology , Disease Models, Animal , Female , Genetic Association Studies , Inflammasomes/immunology , Interferon-beta/metabolism , Interferon-beta/pharmacology , Male , Mice , Mice, Inbred NOD , Molecular Sequence Data , Protein Interaction Domains and Motifs , Sequence Alignment
3.
Methods Mol Biol ; 1278: 233-8, 2015.
Article in English | MEDLINE | ID: mdl-25859953

ABSTRACT

The stoichiometry of a protein complex can be calculated from an accurate measurement of the complex's molecular weight. Multiangle laser light scattering in combination with size exclusion chromatography and interferometric refractometry provides a powerful means for determining the molecular weights of proteins and protein complexes. In contrast to conventional size exclusion chromatography and analytical centrifugation, measurements do not rely on the use of molecular weight standards and are not affected by the shape of the proteins. The technique is based on the direct relationship between the amount of light scattered by a protein in solution, and the product of its concentration and molecular weight. A typical experimental configuration includes a size exclusion column to fractionate the sample, a light scattering detector to measure scattered light, and an interferometric refractometer to measure protein concentration. The determination of the molecular weight of an anthrax toxin complex will be used to illustrate how multiangle laser light scattering can be used to determine the stoichiometry of protein complexes.


Subject(s)
Dynamic Light Scattering , Multiprotein Complexes/chemistry , Proteins/chemistry , Refractometry , Chromatography, Gel , Interferometry , Molecular Weight , Solutions/chemistry
4.
Infect Immun ; 82(9): 3697-703, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24935976

ABSTRACT

Pattern recognition receptors monitor for signs of infection or cellular dysfunction and respond to these events by initiating an immune response. NLRP1B is a receptor that upon activation recruits multiple copies of procaspase-1, which promotes cytokine processing and a proinflammatory form of cell death termed pyroptosis. NLRP1B detects anthrax lethal toxin when the toxin cleaves an amino-terminal fragment from the protein. In addition, NLRP1B is activated when cells are deprived of glucose or treated with metabolic inhibitors, but the mechanism by which the resulting reduction in cytosolic ATP is sensed by NLRP1B is unknown. Here, we addressed whether these two activating signals of NLRP1B converge on a common sensing system. We show that an NLRP1B mutant lacking the amino-terminal region exhibits some spontaneous activity and fails to be further activated by lethal toxin. This mutant was still activated in cells depleted of ATP, however, indicating that the amino-terminal region is not the sole sensing domain of NLRP1B. Mutagenesis of the leucine-rich repeat domain of NLRP1B provided evidence that this domain is involved in autoinhibition of the receptor, but none of the mutants tested was specifically defective at sensing activating signals. Comparison of two alleles of NLRP1B that differed in their response to metabolic inhibitors, but not to lethal toxin, led to the finding that a repeated sequence in the function to find domain (FIIND) that arose from exon duplication facilitated detection of ATP depletion. These results suggest that distinct regions of NLRP1B detect activating signals.


Subject(s)
Anthrax/immunology , Antigens, Bacterial/immunology , Apoptosis Regulatory Proteins/immunology , Bacterial Toxins/immunology , Adenosine Triphosphate/immunology , Anthrax/microbiology , Bacillus anthracis/immunology , Cell Line , Humans , Inflammasomes/immunology , Leucine/immunology , Leucine/metabolism , Receptors, Pattern Recognition/immunology
5.
Angew Chem Int Ed Engl ; 53(31): 8037-40, 2014 Jul 28.
Article in English | MEDLINE | ID: mdl-24706570

ABSTRACT

The design of polyvalent molecules, presenting multiple copies of a specific ligand, represents a promising strategy to inhibit pathogens and toxins. The ability to control independently the valency and the spacing between ligands would be valuable for elucidating structure-activity relationships and for designing potent polyvalent molecules. To that end, we designed monodisperse polypeptide-based polyvalent inhibitors of anthrax toxin in which multiple copies of an inhibitory toxin-binding peptide were separated by flexible peptide linkers. By tuning the valency and linker length, we designed polyvalent inhibitors that were over four orders of magnitude more potent than the corresponding monovalent ligands. This strategy for the rational design of monodisperse polyvalent molecules may not only be broadly applicable for the inhibition of toxins and pathogens, but also for controlling the nanoscale organization of cellular receptors to regulate signaling and the fate of stem cells.


Subject(s)
Bacterial Toxins/antagonists & inhibitors , Peptides/chemistry , Amino Acid Sequence , Antigens, Bacterial , Thermodynamics
6.
Biochemistry ; 53(13): 2166-71, 2014 Apr 08.
Article in English | MEDLINE | ID: mdl-24641616

ABSTRACT

Anthrax toxin protective antigen (PA) binds cellular receptors and self-assembles into oligomeric prepores. A prepore converts to a protein translocating pore after it has been transported to an endosome where the low pH triggers formation of a membrane-spanning ß-barrel channel. Formation of this channel occurs after some PA-receptor contacts are broken to allow pore formation, while others are retained to preserve receptor association. The interaction between PA and anthrax toxin receptor 1 (ANTXR1) is weaker than its interaction with ANTXR2 such that the pH threshold of ANTXR1-mediated pore formation is higher by 1 pH unit. Here we examine receptor-specific differences in toxin binding and pore formation by mutating PA residue G342 that selectively abuts ANTXR2. Mutation of G342 to valine, leucine, isoleucine, or tryptophan increased the amount of PA bound to ANTXR1-expressing cells and decreased the amount of PA bound to ANTXR2-expressing cells. The more conservative G342A mutation did not affect the level of binding to ANTXR2, but ANTXR2-bound PA-G342A prepores exhibited a pH threshold higher than that of wild-type prepores. Mixtures of wild-type PA and PA-G342A were functional in toxicity assays, and the pH threshold of ANTXR2-mediated pore formation was dictated by the relative amounts of the two proteins in the hetero-oligomers. These results suggest that PA subunits within an oligomer do not have to be triggered simultaneously for a productive membrane insertion event to occur.


Subject(s)
Antigens, Bacterial/genetics , Antigens, Bacterial/metabolism , Bacterial Toxins/genetics , Bacterial Toxins/metabolism , Point Mutation/genetics , Receptors, Peptide/metabolism , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Hydrogen-Ion Concentration , Models, Molecular , Receptors, Peptide/chemistry , Structure-Activity Relationship
7.
Infect Immun ; 81(2): 570-9, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23230290

ABSTRACT

The efficacy of the innate immune system depends on its ability to mount an appropriate response to diverse infections and damaging agents. Key components of this system are pattern recognition receptors that detect pathogen-associated and damage-associated molecular patterns (PAMPs and DAMPs). Nlrp1b is a pattern recognition receptor that forms a caspase-1 activation platform, known as an inflammasome, upon sensing the proteolytic activity of anthrax lethal toxin. The activation of caspase-1 leads to the release of proinflammatory cytokines that aid in the clearance of the anthrax infection. Here, we demonstrate that Nlrp1b also becomes activated in cells that are subjected to energy stress caused by metabolic inhibitors or by nutrient deprivation. Glucose starvation and hypoxia were used to correlate the level of cytosolic ATP to the degree of inflammasome activation. Because lowering the ratio of cytosolic ATP to AMP activates the main cellular energy sensor, AMP-activated protein kinase (AMPK), we assessed whether AMPK promoted inflammasome activity by using a combination of small interfering RNA (siRNA) and transfection of a dominant negative AMPK subunit. We found that AMPK promoted inflammasome activity, but activation of AMPK in the absence of ATP depletion was not sufficient for caspase-1-mediated pro-interleukin 1ß (pro-IL-1ß) processing. Finally, we found that mutation of the ATP-binding motif of Nlrp1b caused constitutive activation, suggesting that ATP might inhibit the Nlrp1b inflammasome instead of being required for its assembly.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Adenosine Triphosphate/metabolism , Apoptosis Regulatory Proteins/metabolism , Cytosol/metabolism , Inflammasomes/metabolism , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adenosine Monophosphate/genetics , Adenosine Monophosphate/metabolism , Adenosine Triphosphate/genetics , Amino Acid Motifs , Apoptosis Regulatory Proteins/genetics , Caspase 1/genetics , Caspase 1/metabolism , Cell Hypoxia/physiology , Cell Line, Tumor , Glucose/metabolism , Humans , Inflammasomes/genetics , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Macrophages/metabolism , Mutation , NLR Proteins , Oxidation-Reduction , Receptors, Pattern Recognition/genetics , Receptors, Pattern Recognition/metabolism , Stress, Physiological/physiology
8.
PLoS Pathog ; 8(4): e1002659, 2012.
Article in English | MEDLINE | ID: mdl-22536155

ABSTRACT

Nlrp1b is a NOD-like receptor that detects the catalytic activity of anthrax lethal toxin and subsequently co-oligomerizes into a pro-caspase-1 activation platform known as an inflammasome. Nlrp1b has two domains that promote oligomerization: a NACHT domain, which is a member of the AAA+ ATPase family, and a poorly characterized Function to Find Domain (FIIND). Here we demonstrate that proteolytic processing within the FIIND generates N-terminal and C-terminal cleavage products of Nlrp1b that remain associated in both the auto-inhibited state and in the activated state after cells have been treated with lethal toxin. Functional significance of cleavage was suggested by the finding that mutations that block processing of Nlrp1b also prevent the ability of Nlrp1b to activate pro-caspase-1. By using an uncleaved mutant of Nlrp1b, we established the importance of cleavage by inserting a heterologous TEV protease site into the FIIND and demonstrating that TEV protease processed this site and induced inflammasome activity. Proteolysis of Nlrp1b was shown to be required for the assembly of a functional inflammasome: a mutation within the FIIND that abolished cleavage had no effect on self-association of a FIIND-CARD fragment, but did reduce the recruitment of pro-caspase-1. Our work indicates that a post-translational modification enables Nlrp1b to function.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Caspase 1/metabolism , Inflammasomes/metabolism , Protein Processing, Post-Translational , Proteolysis , Animals , Apoptosis Regulatory Proteins/genetics , Caspase 1/genetics , Cell Line , Enzyme Activation/genetics , Inflammasomes/genetics , Mice , Protein Structure, Tertiary
9.
Biochemistry ; 51(6): 1249-56, 2012 Feb 14.
Article in English | MEDLINE | ID: mdl-22303962

ABSTRACT

ANTXR1 is a type I membrane protein that binds the protective antigen (PA) component of anthrax toxin. The cytosolic domain of ANTXR1 has a novel actin-binding region that influences the interaction of the ectodomain with PA. Here, we have investigated features of the cytosolic domain of ANTXR1 that reduce the association of the receptor with PA. We mutated a stretch of conserved acidic amino acids adjacent to the actin-binding region and found that the mutation increased the affinity for monomeric actin in vitro. ANTXR1 bearing this mutation exhibited increased association with the cytoskeleton and bound less PA compared to the wild-type receptor, confirming the inverse correlation between the two interactions. To determine whether binding of actin is sufficient to regulate the ectodomain, we replaced the actin-binding region of ANTXR1 with that from the yeast protein abp140 and with the WH2 domain of WAVE2. Although both of these domains bound monomeric actin in vitro, only the sequence from abp140 reduced binding of PA to a hybrid receptor. The actin binding regions of ANTXR1 and abp140, but not the WH2 domain, colocalized with actin stress fibers, which suggested that filamentous actin regulates ANTXR1. Consistent with this notion, disruption of actin filaments using latrunculin A increased the amount of PA bound to cells. This work provides evidence that cytoskeletal dynamics regulate ANTXR1 function.


Subject(s)
Actins/antagonists & inhibitors , Actins/metabolism , Antigens, Bacterial/metabolism , Bacterial Toxins/antagonists & inhibitors , Bacterial Toxins/metabolism , Down-Regulation , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Receptors, Cell Surface/antagonists & inhibitors , Receptors, Cell Surface/metabolism , Actins/genetics , Antigens, Bacterial/chemistry , Antigens, Bacterial/genetics , Bacillus anthracis/genetics , Bacillus anthracis/metabolism , Bacillus anthracis/pathogenicity , Bacterial Toxins/chemistry , Bacterial Toxins/genetics , Cell Line, Tumor , Down-Regulation/genetics , HeLa Cells , Humans , Hydrogen-Ion Concentration , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Membrane Proteins/physiology , Microfilament Proteins , Molecular Dynamics Simulation , Mutation , Neoplasm Proteins/genetics , Protein Binding/genetics , Receptors, Cell Surface/genetics , Receptors, Peptide
10.
Autophagy ; 7(9): 957-65, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21606683

ABSTRACT

Autophagy plays a significant role in innate and adaptive immune responses to microbial infection. Some pathogenic bacteria have developed strategies to evade killing by host autophagy. These include the use of 'camouflage' proteins to block targeting to the autophagy pathway and the use of pore-forming toxins to block autophagosome maturation. However, general inhibition of host autophagy by bacterial pathogens has not been observed to date. Here we demonstrate that bacterial cAMP-elevating toxins from B. anthracis and V. cholera can inhibit host anti-microbial autophagy, including autophagic targeting of S. Typhimurium and latex bead phagosomes. Autophagy inhibition required the cAMP effector protein kinase A. Formation of autophagosomes in response to rapamycin and the endogenous turnover of peroxisomes was also inhibited by cAMP-elevating toxins. These findings demonstrate that cAMP-elevating toxins, representing a large group of bacterial virulence factors, can inhibit host autophagy to suppress immune responses and modulate host cell physiology.


Subject(s)
Autophagy/drug effects , Bacterial Toxins/pharmacology , Cyclic AMP/metabolism , Animals , Cyclic AMP-Dependent Protein Kinases/metabolism , Enzyme Activation/drug effects , HeLa Cells , Humans , Intracellular Space/drug effects , Intracellular Space/microbiology , Macrophages/cytology , Macrophages/drug effects , Macrophages/microbiology , Mice , Receptors, IgG/metabolism , Salmonella typhimurium/drug effects , Salmonella typhimurium/growth & development , Salmonella typhimurium/physiology
11.
Biomacromolecules ; 12(3): 791-6, 2011 Mar 14.
Article in English | MEDLINE | ID: mdl-21302959

ABSTRACT

The design of polyvalent molecules, consisting of multiple copies of a biospecific ligand attached to a suitable scaffold, represents a promising approach to inhibit pathogens and oligomeric microbial toxins. Despite the increasing interest in structure-based drug design, few polyvalent inhibitors based on this approach have shown efficacy in vivo. Here we demonstrate the structure-based design of potent biospecific heptavalent inhibitors of anthrax lethal toxin. Specifically, we illustrate the ability to design potent polyvalent ligands by matching the pattern of binding sites on the biological target. We used a combination of experimental studies based on mutagenesis and computational docking studies to identify the binding site for an inhibitory peptide on the heptameric subunit of anthrax toxin. We developed an approach based on copper-catalyzed azide-alkyne cycloaddition (click-chemistry) to facilitate the attachment of seven copies of the inhibitory peptide to a ß-cyclodextrin core via a polyethylene glycol linker of an appropriate length. The resulting heptavalent inhibitors neutralized anthrax lethal toxin both in vitro and in vivo and showed appreciable stability in serum. Given the inherent biocompatibility of cyclodextrin and polyethylene glycol, these potent well-defined heptavalent inhibitors show considerable promise as anthrax antitoxins.


Subject(s)
Antitoxins/chemistry , Bacterial Toxins/antagonists & inhibitors , Cyclodextrins/chemistry , Antigens, Bacterial , Binding Sites , Drug Design , Drug Stability , Peptides/chemistry , Peptides/pharmacology , Structure-Activity Relationship
12.
FEBS J ; 277(1): 119-27, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19922472

ABSTRACT

Anthrax lethal toxin triggers death in some cell types, such as macrophages, and causes a variety of cellular dysfunctions in others. Collectively, these effects dampen the innate and adaptive immune systems to allow Bacillus anthracis to survive and proliferate in the mammalian host. The diverse effects caused by the toxin have in part been attributed to its interference with signaling pathways in target cells. Lethal factor (LF) is the proteolytic component of the toxin, and cleaves six members of the mitogen-activated protein kinase kinase family after being delivered to the cytosol by the cell-binding component of the toxin, protective antigen. The effect of cleaving these mitogen-activated protein kinase kinases is to interfere with extracellular signal-related kinase (ERK), p38 and c-Jun N-terminal kinase signaling. Here, we characterized an LF mutant, LF-K518E/E682G, that was defective at causing pyroptosis in RAW 264.7 cells and at activating the Nlrp1b inflammasome in a heterologous expression system. LF-K518E/E682G did not exhibit an overall impairment of function, however, because it was able to downregulate the ERK pathway, but not the p38 or c-Jun N-terminal kinase pathways. Furthermore, LF-K518E/E682G efficiently killed melanoma cells, which were shown previously to undergo apoptosis in response to lethal toxin or to pharmacological inhibition of the ERK pathway. Our results suggest that LF-K518E/E682G is defective at cleaving a substrate involved in the activation of the Nlrp1b inflammasome.


Subject(s)
Antigens, Bacterial/genetics , Antigens, Bacterial/toxicity , Apoptosis/drug effects , Bacillus anthracis/genetics , Bacillus anthracis/pathogenicity , Bacterial Toxins/genetics , Bacterial Toxins/toxicity , Animals , Antigens, Bacterial/chemistry , Antigens, Bacterial/immunology , Apoptosis/immunology , Bacillus anthracis/immunology , Bacterial Toxins/chemistry , Bacterial Toxins/immunology , Cell Line , Cell Line, Tumor , Host-Pathogen Interactions/immunology , Host-Pathogen Interactions/physiology , Humans , MAP Kinase Signaling System/drug effects , Mice , Models, Molecular , Mutagenesis , Mutation , Protein Structure, Tertiary , Virulence/genetics
13.
J Biol Chem ; 285(3): 2120-9, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-19858192

ABSTRACT

Anthrax lethal toxin (LeTx) is a virulence factor secreted by Bacillus anthracis and has direct cytotoxic effects on most cells once released into the cytoplasm. The cytoplasmic delivery of the proteolytically active component of LeTx, lethal factor (LF), is carried out by the transporter component, protective antigen, which interacts with either of two known surface receptors known as anthrax toxin receptor (ANTXR) 1 and 2. We found that the cytoplasmic delivery of LF by ANTXR2 was mediated by cathepsin B (CTSB) and required lysosomal fusion with LeTx-containing endosomes. Also, binding of protective antigen to ANXTR1 or -2 triggered autophagy, which facilitated the cytoplasmic delivery of ANTXR2-associated LF. We found that whereas cells treated with the membrane-permeable CTSB inhibitor CA074-Me- or CTSB-deficient cells had no defect in fusion of LC3-containing autophagic vacuoles with lysosomes, autophagic flux was significantly delayed. These results suggested that the ANTXR2-mediated cytoplasmic delivery of LF was enhanced by CTSB-dependent autophagic flux.


Subject(s)
Antigens, Bacterial/metabolism , Autophagy , Bacterial Toxins/metabolism , Cathepsin B/metabolism , Cytoplasm/metabolism , Endocytosis , Receptors, Peptide/metabolism , Animals , Cathepsin B/antagonists & inhibitors , Cell Line , Cytoplasm/drug effects , Dipeptides/pharmacology , Endocytosis/drug effects , Endosomes/drug effects , Endosomes/metabolism , Humans , Lysosomes/drug effects , Lysosomes/metabolism , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Mice , Protease Inhibitors/pharmacology
14.
Cell Microbiol ; 12(4): 557-68, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-19995385

ABSTRACT

Anthrax lethal toxin (LeTx) is composed of protective antigen (PA) and lethal factor (LF) - PA is the receptor-binding moiety and LF is a protease that cleaves mitogen-activated protein kinase kinases (MAPKKs). LeTx subverts the immune response to Bacillus anthracis in several ways, such as downregulating interleukin-8 (IL-8) by increasing the rate of IL-8 mRNA degradation. Many transcripts are regulated through cis-acting elements that bind proteins that either impede or promote degradation. Some of these RNA-binding proteins are regulated by MAPKs and previous work has demonstrated that interfering with MAPK signalling decreases the half-life of IL-8 mRNA. Here, we have localized a segment within the IL-8 3' untranslated region responsible for LeTx-induced transcript destabilization and show that this is caused by inhibition of the p38, ERK and JNK pathways. TTP, an RNA-binding protein involved in IL-8 mRNA decay, became hypophosphorylated in LeTx-treated cells and knock-down of TTP prevented LeTx from destabilizing the IL-8 transcript. Cells that were treated with LeTx exhibited increased localization of TTP to Processing bodies, which are structures that accumulate transcripts targeted for degradation. We furthermore observed that LeTx promoted the formation of Processing bodies, revealing a link between the toxin and a major mRNA decay pathway.


Subject(s)
Antigens, Bacterial/toxicity , Bacillus anthracis/immunology , Bacillus anthracis/pathogenicity , Bacterial Toxins/toxicity , Interleukin-8/biosynthesis , RNA Stability , Tristetraprolin/metabolism , 3' Untranslated Regions , Antigens, Bacterial/metabolism , Bacterial Toxins/metabolism , Cell Line , Gene Knockdown Techniques , Humans , Interleukin-8/genetics , Phosphorylation , Tristetraprolin/antagonists & inhibitors
15.
Biochemistry ; 48(44): 10577-81, 2009 Nov 10.
Article in English | MEDLINE | ID: mdl-19817382

ABSTRACT

The protective antigen component of anthrax toxin binds the I domain of the anthrax toxin receptors, ANTXR1 and ANTXR2, in a manner akin to how integrins bind their ligands. The I domains of integrins and ANTXR1 both have high- and low-affinity conformations, and the cytosolic tails of these receptors associate with the actin cytoskeleton. The association of ANTXR1 with the cytoskeleton correlates with weakened binding to PA, although a mechanistic explanation for this observation is lacking. Here, we identified a segment in the cytoplasmic tail of ANTXR1 required for its association with the cytoskeleton. We synthesized a 60-mer peptide based on this segment and demonstrated a direct interaction between the peptide and beta-actin, indicating that in contrast to integrins, ANTXR1 does not use an adaptor to bind the cytoskeleton. This peptide orders actin filaments into arrays, demonstrating an actin bundling activity that is novel for a membrane protein.


Subject(s)
Actins/metabolism , Cytoskeleton/metabolism , Neoplasm Proteins/metabolism , Receptors, Cell Surface/metabolism , Base Sequence , DNA Primers , Electrophoresis, Polyacrylamide Gel , HeLa Cells , Humans , Microfilament Proteins , Microscopy, Electron
16.
Infect Immun ; 77(10): 4455-62, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19651869

ABSTRACT

Anthrax lethal toxin causes macrophages and dendritic cells from some mouse strains to undergo caspase-1-dependent cell death. Central to this process is the NOD-like receptor Nlrp1b (Nalp1b), which detects intoxication and then self-associates to form a complex, termed an inflammasome, that is capable of activating the procaspase-1 zymogen. The nature of the signal detected directly by Nlrp1b is not known, and the mechanisms of inflammasome assembly are poorly understood. Here, we demonstrate that transfection of human fibroblasts with plasmids encoding murine Nlrp1b and procaspase-1 was sufficient to confer susceptibility to lethal toxin-mediated death on the cells. As has been observed in murine macrophages, the enzymatic activities of lethal toxin and the proteasome were both required for activation of the Nlrp1b inflammasome and this activation led to prointerleukin-1 beta processing. Release of interleukin-1beta from cells was not dependent on cell lysis, as its secretion was not affected by an osmoprotectant that prevented the appearance of lactate dehydrogenase in the culture medium. We generated constitutively active mutants of Nlrp1b by making amino-terminal deletions to the protein and observed that the ability to activate procaspase-1 was dependent on the CARD domain, which bound procaspase-1, and a region adjacent to the CARD domain that promoted self-association. Our results demonstrate that lethal toxin can activate Nlrp1b in a nonmyeloid cell line and are consistent with work that suggests that activation induces proximity of procaspase-1.


Subject(s)
Antigens, Bacterial/toxicity , Apoptosis Regulatory Proteins/physiology , Bacterial Toxins/toxicity , Fibroblasts/drug effects , Animals , Caspase 1/genetics , Caspase 1/physiology , Cell Survival , Gene Expression , Humans , Mice , Plasmids , Transfection
17.
Infect Immun ; 77(1): 52-9, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18936178

ABSTRACT

The protective antigen (PA) component of anthrax toxin binds the I domain of the receptor ANTXR1. Integrin I domains convert between open and closed conformations that bind ligand with high and low affinities, respectively; this process is regulated by signaling from the cytoplasmic domains. To assess whether intracellular signals might influence the interaction between ANTXR1 and PA, we compared two splice variants of ANTXR1 that differ only in their cytoplasmic domains. We found that cells expressing ANTXR1 splice variant 1 (ANTXR1-sv1) bound markedly less PA than did cells expressing a similar level of the shorter splice variant ANTXR1-sv2. ANTXR1-sv1 but not ANTXR1-sv2 associated with the actin cytoskeleton, although disruption of the cytoskeleton did not affect binding of ANTXR-sv1 to PA. Introduction of a cytoplasmic domain missense mutation found in the related receptor ANTXR2 in a patient with juvenile hyaline fibromatosis impaired actin association and increased binding of PA to ANTXR1-sv1. These results suggest that ANTXR1 has two affinity states that may be modulated by cytoplasmic signals.


Subject(s)
Antigens, Bacterial/metabolism , Bacterial Toxins/metabolism , Receptors, Peptide/metabolism , Amino Acid Substitution/genetics , Animals , CHO Cells , Cricetinae , Cricetulus , Mutagenesis, Site-Directed , Mutation, Missense , Protein Binding , Protein Structure, Tertiary/genetics , Receptors, Peptide/genetics
19.
PLoS One ; 3(1): e1421, 2008 Jan 09.
Article in English | MEDLINE | ID: mdl-18183301

ABSTRACT

BACKGROUND: Bacillus anthracis is the bacterium responsible for causing anthrax. The ability of B. anthracis to cause disease is dependent on a secreted virulence factor, lethal toxin, that promotes survival of the bacteria in the host by impairing the immune response. A well-studied effect of lethal toxin is the killing of macrophages, although the molecular mechanisms involved have not been fully characterized. METHODOLOGY/PRINCIPAL FINDINGS: Here, we demonstrate that celastrol, a quinone methide triterpene derived from a plant extract used in herbal medicine, inhibits lethal toxin-induced death of RAW264.7 murine macrophages. Celastrol did not prevent cleavage of mitogen activated protein kinase kinase 1, a cytosolic target of the toxin, indicating that it did not inhibit the uptake or catalytic activity of lethal toxin. Surprisingly, celastrol conferred almost complete protection when it was added up to 1.5 h after intoxication, indicating that it could rescue cells in the late stages of intoxication. Since the activity of the proteasome has been implicated in intoxication using other pharmacological agents, we tested whether celastrol blocked proteasome activity. We found that celastrol inhibited the proteasome-dependent degradation of proteins in RAW264.7 cells, but only slightly inhibited proteasome-mediated cleavage of fluorogenic substrates in vitro. Furthermore, celastrol blocked stimulation of IL-18 processing, indicating that celastrol acted upstream of inflammasome activation. CONCLUSIONS/SIGNIFICANCE: This work identifies celastrol as an inhibitor of lethal toxin-mediated macrophage lysis and suggests an inhibitory mechanism involving inhibition of the proteasome pathway.


Subject(s)
Bacterial Toxins/antagonists & inhibitors , Macrophages/drug effects , Triterpenes/pharmacology , Animals , Antigens, Bacterial/toxicity , Bacterial Toxins/toxicity , Blotting, Western , Cell Line , Hydrolysis , Macrophages/cytology , Mice , Pentacyclic Triterpenes , Proteasome Endopeptidase Complex/metabolism , Tripterygium/chemistry
20.
J Polym Sci A Polym Chem ; 46(21): 7246-7257, 2008 Sep 25.
Article in English | MEDLINE | ID: mdl-19855852

ABSTRACT

We describe the synthesis of activated homopolymers and copolymers of controlled molecular weight based on the controlled radical polymerization of N-acryloyloxysuccinimide (NAS) by reversible addition fragmentation chain transfer (RAFT). We synthesized activated homopolymers in a range of molecular weights with polydispersities between 1 and 1.2. The attachment of an inhibitory peptide to the activated polymer backbone yielded a potent controlled molecular weight polyvalent inhibitor of anthrax toxin. To provide greater control over the placement of the peptides along the polymer backbone, we also used a semi-batch copolymerization method to synthesize copolymers of NAS and acrylamide (AAm). This approach enabled the synthesis of copolymers with control over the placement of peptide-reactive NAS monomers along an inert backbone; subsequent functionalization of NAS with peptide yielded well-defined polyvalent anthrax toxin inhibitors that differed in their potencies. These strategies for controlling molecular weight, ligand density, and ligand placement will be broadly applicable for designing potent polyvalent inhibitors for a variety of pathogens and toxins, and for elucidating structure-activity relationships in these systems.

SELECTION OF CITATIONS
SEARCH DETAIL
...