Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Perm J ; 242020.
Article in English | MEDLINE | ID: mdl-33202215

ABSTRACT

Furin is a protease that is ubiquitous in mammalian metabolism. One of the innovations that make sudden acute respiratory syndrome-coronavirus 2 (SARS-CoV-2) more infectious than its ancestor viruses is the addition of a furin cleavage site. Conditions associated with elevated furin levels, including diabetes, obesity, and hypertension, overlap greatly with vulnerability to the severe form of coronavirus disease 2019 (COVID-19). We suggest that diet and lifestyle modifications that reduce the associated comorbidities may prevent the development of severe COVID-19 by, in part, lowering circulating furin levels. Likewise, natural and pharmaceutical inhibitors of furin may be candidate prophylactic interventions or, if used early in the COVID-19, may prevent the development of critical symptoms.


Subject(s)
Anthrax/blood , COVID-19/blood , Diabetes Mellitus/blood , Furin/blood , Hypertension/blood , Severe acute respiratory syndrome-related coronavirus/metabolism , Anthrax/enzymology , COVID-19/enzymology , Diabetes Mellitus/enzymology , Humans , Hypertension/enzymology , Severe acute respiratory syndrome-related coronavirus/enzymology
2.
Nat Microbiol ; 5(12): 1464-1471, 2020 12.
Article in English | MEDLINE | ID: mdl-32895527

ABSTRACT

Anthrax lethal toxin (LT), produced by Bacillus anthracis, comprises a receptor-binding moiety, protective antigen and the lethal factor (LF) protease1,2. Although LF is known to cleave mitogen-activated protein kinase kinases (MEKs/MKKs) and some variants of the NLRP1 inflammasome sensor, targeting of these pathways does not explain the lethality of anthrax toxin1,2. Here we report that the regulatory subunits of phosphoinositide-3 kinase (PI3K)-p85α (PIK3R1) and p85ß (PIK3R2)3,4-are substrates of LF. Cleavage of these proteins in a proline-rich region between their N-terminal Src homology and Bcr homology domains disrupts homodimer formation and impacts PI3K signalling. Mice carrying a mutated p85α that cannot be cleaved by LF show a greater resistance to anthrax toxin challenge. The LF(W271A) mutant cleaves p85α with lower efficiency and is non-toxic to mice but can regain lethality when combined with PI3K pathway inhibitors. We provide evidence that LF targets two signalling pathways that are essential for growth and metabolism and that the disabling of both pathways is likely necessary for lethal anthrax infection.


Subject(s)
Anthrax/enzymology , Antigens, Bacterial/metabolism , Antigens, Bacterial/toxicity , Bacillus anthracis/enzymology , Bacillus anthracis/metabolism , Bacterial Toxins/metabolism , Bacterial Toxins/toxicity , Class Ia Phosphatidylinositol 3-Kinase/metabolism , Peptide Hydrolases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Amino Acid Motifs , Animals , Anthrax/genetics , Anthrax/microbiology , Class Ia Phosphatidylinositol 3-Kinase/chemistry , Class Ia Phosphatidylinositol 3-Kinase/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Peptide Hydrolases/genetics , Phosphatidylinositol 3-Kinases/chemistry , Phosphatidylinositol 3-Kinases/genetics
3.
Med Sci Monit ; 20: 1151-4, 2014 Jul 06.
Article in English | MEDLINE | ID: mdl-24997584

ABSTRACT

BACKGROUND: Adenosine deaminase (ADA) activity has been discovered in several inflammatory conditions; however, there are no data associated with cutaneous anthrax. The aim of this study was to investigate serum ADA activity in patients with cutaneous anthrax. MATERIAL AND METHODS: Sixteen patients with cutaneous anthrax and 17 healthy controls were enrolled. We measured ADA activity; peripheral blood leukocyte, lymphocyte, neutrophil, and monocyte counts; erythrocyte sedimentation rate; and C reactive protein levels. RESULTS: Serum ADA activity was significantly higher in patients with cutaneous anthrax than in the controls (p<0.001). A positive correlation was observed between ADA activity and lymphocyte counts (r=0.589, p=0.021) in the patient group. CONCLUSIONS: This study suggests that serum ADA could be used as a biochemical marker in cutaneous anthrax.


Subject(s)
Adenosine Deaminase/blood , Anthrax/blood , Anthrax/enzymology , Skin Diseases, Bacterial/blood , Skin Diseases, Bacterial/enzymology , Adult , Case-Control Studies , Demography , Female , Humans , Male
4.
PLoS One ; 9(6): e100532, 2014.
Article in English | MEDLINE | ID: mdl-24945934

ABSTRACT

Gastrointestinal (GI) anthrax results from the ingestion of Bacillus anthracis. Herein, we investigated the pathogenesis of GI anthrax in animals orally infected with toxigenic non-encapsulated B. anthracis Sterne strain (pXO1+ pXO2-) spores that resulted in rapid animal death. B. anthracis Sterne induced significant breakdown of intestinal barrier function and led to gut dysbiosis, resulting in systemic dissemination of not only B. anthracis, but also of commensals. Disease progression significantly correlated with the deterioration of innate and T cell functions. Our studies provide critical immunologic and physiologic insights into the pathogenesis of GI anthrax infection, whereupon cleavage of mitogen-activated protein kinases (MAPKs) in immune cells may play a central role in promoting dysfunctional immune responses against this deadly pathogen.


Subject(s)
Anthrax/immunology , Anthrax/microbiology , Bacillus anthracis/immunology , Colon/immunology , Dysbiosis/immunology , Dysbiosis/microbiology , Gastrointestinal Diseases/immunology , Gastrointestinal Diseases/microbiology , Immune Tolerance , Animals , Anthrax/enzymology , Anthrax/pathology , Colon/microbiology , Colon/pathology , Dysbiosis/pathology , Epithelium/immunology , Epithelium/microbiology , Epithelium/pathology , Gastrointestinal Diseases/enzymology , Gastrointestinal Diseases/pathology , Immunity, Innate , Mice , Mitogen-Activated Protein Kinases/metabolism , T-Lymphocytes/immunology
5.
J Biol Chem ; 288(32): 23458-72, 2013 Aug 09.
Article in English | MEDLINE | ID: mdl-23720780

ABSTRACT

Toxins play a major role in the pathogenesis of Bacillus anthracis by subverting the host defenses. However, besides toxins, B. anthracis expresses effector proteins, whose role in pathogenesis are yet to be investigated. Here we present that suppressor-of-variegation, enhancer-of-zeste, trithorax protein from B. anthracis (BaSET) methylates human histone H1, resulting in repression of NF-κB functions. Notably, BaSET is secreted and undergoes nuclear translocation to enhance H1 methylation in B. anthracis-infected macrophages. Compared with wild type Sterne, delayed growth kinetics and altered septum formation were observed in the BaSET knock-out (BaΔSET) bacilli. Uncontrolled BaSET expression during complementation of the BaSET gene in BaΔSET partially restored growth during stationary phase but resulted in substantially shorter bacilli throughout the growth cycle. Importantly, in contrast to Sterne, the BaΔSET B. anthracis is avirulent in a lethal murine bacteremia model of infection. Collectively, BaSET is required for repression of host transcription as well as proper B. anthracis growth, making it a potentially unique virulence determinant.


Subject(s)
Anthrax/enzymology , Bacillus anthracis , Bacterial Proteins/biosynthesis , Epigenesis, Genetic , Macrophages/metabolism , NF-kappa B/metabolism , Protein Methyltransferases/biosynthesis , Transcription, Genetic , Virulence Factors/biosynthesis , Animals , Anthrax/genetics , Anthrax/pathology , Bacillus anthracis/enzymology , Bacillus anthracis/genetics , Bacillus anthracis/pathogenicity , Bacterial Proteins/genetics , Gene Expression Regulation, Bacterial/physiology , HeLa Cells , Humans , Macrophages/microbiology , Macrophages/pathology , Mice , NF-kappa B/genetics , Protein Methyltransferases/genetics , Virulence Factors/genetics
6.
Biochim Biophys Acta ; 1834(1): 46-52, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22999981

ABSTRACT

BACKGROUND: Bacterial resistance to antibiotic therapies is increasing and new treatment options are badly needed. There is an overlap between these resistant bacteria and organisms classified as likely bioterror weapons. For example, Bacillus anthracis is innately resistant to the anti-folate trimethoprim due to sequence changes found in the dihydrofolate reductase enzyme. Development of new inhibitors provides an opportunity to enhance the current arsenal of anti-folate antibiotics while also expanding the coverage of the anti-folate class. METHODS: We have characterized inhibitors of B. anthracis dihydrofolate reductase by measuring the K(i) and MIC values and calculating the energetics of binding. This series contains a core diaminopyrimidine ring, a central dimethoxybenzyl ring, and a dihydrophthalazine moiety. We have altered the chemical groups extended from a chiral center on the dihydropyridazine ring of the phthalazine moiety. The interactions for the most potent compounds were visualized by X-ray structure determination. RESULTS: We find that the potency of individual enantiomers is divergent with clear preference for the S-enantiomer, while maintaining a high conservation of contacts within the binding site. The preference for enantiomers seems to be predicated largely by differential interactions with protein residues Leu29, Gln30 and Arg53. CONCLUSIONS: These studies have clarified the activity of modifications and of individual enantiomers, and highlighted the role of the less-active R-enantiomer in effectively diluting the more active S-enantiomer in racemic solutions. This directly contributes to the development of new antimicrobials, combating trimethoprim resistance, and treatment options for potential bioterrorism agents.


Subject(s)
Bacillus anthracis/enzymology , Bacterial Proteins/antagonists & inhibitors , Drug Design , Folic Acid Antagonists/chemistry , Molecular Docking Simulation , Tetrahydrofolate Dehydrogenase/chemistry , Anthrax/drug therapy , Anthrax/enzymology , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Crystallography, X-Ray , Folic Acid Antagonists/therapeutic use , Structure-Activity Relationship , Tetrahydrofolate Dehydrogenase/metabolism
7.
Biochem Biophys Res Commun ; 430(1): 125-30, 2013 Jan 04.
Article in English | MEDLINE | ID: mdl-23178574

ABSTRACT

Bacillus anthracis, a causative agent of anthrax, is able to germinate and survive within macrophages. A recent study suggested that B. anthracis-derived nitric oxide (bNO) is a key aspect of bacterial defense that protects bacterial DNA from oxidative burst in the macrophages. However, the virulent effect of bNO in host cells has not been investigated. Here, we report that bNO contributes macrophage killing by S-nitrosylation of bioenergetic-relating proteins within mitochondria. Toxigenic Sterne induces expression of the bnos gene and produces bNO during early stage of infection. Nitroso-proteomic analysis coupled with a biotin-switch technique demonstrated that toxigenic infection induces protein S-nitrosylation in B. anthracis-susceptible RAW264.7. For each target enzyme tested (complex I, complex III and complex IV), infection by B. anthracis Sterne caused enzyme inhibition. Nω-nitro-L-arginine methyl ester, a NO synthase inhibitor, reduced S-nitrosylation and partially restored cell viability evaluated by intracellular ATP levels in macrophages. Our data suggest that bNO leads to energy depletion driven by impaired mitochondrial bioenergetic machinery that ultimately contributes to macrophage death. This novel mechanism of anthrax pathogenesis may offer specific approach to the development of therapeutics.


Subject(s)
Anthrax/immunology , Apoptosis/immunology , Bacillus anthracis/pathogenicity , Macrophages/microbiology , Mitochondria/microbiology , Nitric Oxide Synthase Type I/metabolism , Nitric Oxide/metabolism , Animals , Anthrax/enzymology , Anthrax/microbiology , Bacillus anthracis/metabolism , Cell Line , Electron Transport Complex I/metabolism , Electron Transport Complex III/metabolism , Electron Transport Complex IV/metabolism , Gene Expression , Macrophages/enzymology , Mice , Mitochondria/enzymology , Nitric Oxide Synthase Type I/genetics
8.
J Biol Chem ; 286(37): 32586-92, 2011 Sep 16.
Article in English | MEDLINE | ID: mdl-21768086

ABSTRACT

Past anthrax attacks in the United States have highlighted the need for improved measures against bioweapons. The virulence of anthrax stems from the shielding properties of the Bacillus anthracis poly-γ-d-glutamic acid capsule. In the presence of excess CapD, a B. anthracis γ-glutamyl transpeptidase, the protective capsule is degraded, and the immune system can successfully combat infection. Although CapD shows promise as a next generation protein therapeutic against anthrax, improvements in production, stability, and therapeutic formulation are needed. In this study, we addressed several of these problems through computational protein engineering techniques. We show that circular permutation of CapD improved production properties and dramatically increased kinetic thermostability. At 45 °C, CapD was completely inactive after 5 min, but circularly permuted CapD remained almost entirely active after 30 min. In addition, we identify an amino acid substitution that dramatically decreased transpeptidation activity but not hydrolysis. Subsequently, we show that this mutant had a diminished capsule degradation activity, suggesting that CapD catalyzes capsule degradation through a transpeptidation reaction with endogenous amino acids and peptides in serum rather than hydrolysis.


Subject(s)
Anthrax/drug therapy , Bacillus anthracis/enzymology , Bacterial Proteins/chemistry , Computer Simulation , Protein Engineering , gamma-Glutamyltransferase/chemistry , Amino Acid Sequence , Animals , Anthrax/enzymology , Anthrax/genetics , Bacillus anthracis/genetics , Bacterial Capsules/chemistry , Bacterial Capsules/genetics , Bacterial Capsules/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/therapeutic use , Humans , Hydrolysis , Models, Molecular , Molecular Sequence Data , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/therapeutic use , gamma-Glutamyltransferase/genetics , gamma-Glutamyltransferase/metabolism , gamma-Glutamyltransferase/therapeutic use
9.
Biochim Biophys Acta ; 1804(6): 1369-75, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20170757

ABSTRACT

Acetohydroxyacid synthase (AHAS), a potential target for antimicrobial agents, catalyzes the first common step in the biosynthesis of the branched-chain amino acids. The genes of both catalytic and regulatory subunits of AHAS from Bacillus anthracis (Bantx), a causative agent of anthrax, were cloned, overexpressed in Escherichia coli, and purified to homogeneity. To develop novel anti-anthracis drugs that inhibit AHAS, a chemical library was screened, and four chemicals, AVS2087, AVS2093, AVS2387, and AVS2236, were identified as potent inhibitors of catalytic subunit with IC(50) values of 1.0 +/- 0.02, 1.0 +/- 0.04, 2.1 +/- 0.12, and 2.0 +/- 0.08 microM, respectively. Further, these four chemicals also showed strong inhibition against reconstituted AHAS with IC(50) values of 0.05 +/- 0.002, 0.153 +/- 0.004, 1.30 +/- 0.10, and 1.29 +/- 0.40 microM, respectively. The basic scaffold of the AVS group consists of 1-pyrimidine-2-yl-1H-[1,2,4]triazole-3-sulfonamide. The potent inhibitor, AVS2093 showed the lowest binding energy, -8.52 kcal/mol and formed a single hydrogen bond with a distance of 1.973 A. As the need for novel antibiotic classes to combat bacterial drug resistance increases, the screening of new compounds that act against Bantx-AHAS shows that AHAS is a good target for new anti-anthracis drugs.


Subject(s)
Aldehyde-Ketone Transferases/antagonists & inhibitors , Aldehyde-Ketone Transferases/chemistry , Anti-Bacterial Agents/chemistry , Bacillus anthracis/enzymology , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/chemistry , Enzyme Inhibitors/chemistry , Pyrimidines/chemistry , Aldehyde-Ketone Transferases/genetics , Aldehyde-Ketone Transferases/metabolism , Anthrax/drug therapy , Anthrax/enzymology , Anti-Bacterial Agents/therapeutic use , Catalytic Domain , Enzyme Inhibitors/therapeutic use , Hydrogen Bonding , Protein Binding , Pyrimidines/therapeutic use , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
10.
Toxins (Basel) ; 2(5): 1038-53, 2010 05.
Article in English | MEDLINE | ID: mdl-22069624

ABSTRACT

Anthrax is caused by the gram-positive bacterium Bacillus anthracis. The pathogenesis of this disease is dependent on the presence of two binary toxins, edema toxin (EdTx) and lethal toxin (LeTx). LeTx, the major virulence factor contributing to anthrax, contains the effector moiety lethal factor (LF), a zinc-dependent metalloprotease specific for targeting mitogen-activated protein kinase kinases. This review will focus on the protease-specific activity and function of LF, and will include a discussion on the implications and consequences of this activity, both in terms of anthrax disease, and how this activity can be exploited to gain insight into other pathologic conditions.


Subject(s)
Antigens, Bacterial/metabolism , Bacterial Toxins/metabolism , Metalloproteases/metabolism , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Zinc/metabolism , Animals , Anthrax/enzymology , Bacillus anthracis/enzymology , Bacillus anthracis/pathogenicity , Humans
11.
J Biol Chem ; 285(8): 5188-95, 2010 Feb 19.
Article in English | MEDLINE | ID: mdl-19948665

ABSTRACT

Bacillus anthracis is a gram-positive spore-forming bacterium that causes anthrax. With the increased threat of anthrax in biowarfare, there is an urgent need to characterize new antimicrobial targets from B. anthracis. One such target is dihydrodipicolinate synthase (DHDPS), which catalyzes the committed step in the pathway yielding meso-diaminopimelate and lysine. In this study, we employed CD spectroscopy to demonstrate that the thermostability of DHDPS from B. anthracis (Ba-DHDPS) is significantly enhanced in the presence of the substrate, pyruvate. Analytical ultracentrifugation studies show that the tetramer-dimer dissociation constant of the enzyme is 3-fold tighter in the presence of pyruvate compared with the apo form. To examine the significance of this substrate-mediated stabilization phenomenon, a dimeric mutant of Ba-DHDPS (L170E/G191E) was generated and shown to have markedly reduced activity compared with the wild-type tetramer. This demonstrates that the substrate, pyruvate, stabilizes the active form of the enzyme. We next determined the high resolution (2.15 A) crystal structure of Ba-DHDPS in complex with pyruvate (3HIJ) and compared this to the apo structure (1XL9). Structural analyses show that there is a significant (91 A(2)) increase in buried surface area at the tetramerization interface of the pyruvate-bound structure. This study describes a new mechanism for stabilization of the active oligomeric form of an antibiotic target from B. anthracis and reveals an "Achilles heel" that can be exploited in structure-based drug design.


Subject(s)
Anthrax/enzymology , Bacillus anthracis/enzymology , Bacterial Proteins/chemistry , Hydro-Lyases/chemistry , Pyruvic Acid/chemistry , Amino Acid Substitution , Anthrax/drug therapy , Anthrax/genetics , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/therapeutic use , Bacillus anthracis/genetics , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biological Warfare Agents , Diaminopimelic Acid/chemistry , Diaminopimelic Acid/metabolism , Drug Design , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/therapeutic use , Hydro-Lyases/antagonists & inhibitors , Hydro-Lyases/genetics , Hydro-Lyases/metabolism , Lysine/chemistry , Lysine/genetics , Lysine/metabolism , Mutation, Missense , Protein Structure, Quaternary/physiology , Pyruvic Acid/metabolism
12.
Mol Aspects Med ; 30(6): 431-8, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19665472

ABSTRACT

The anthrax lethal factor is a multi-domain protein toxin released by Bacillus anthracis which enters cells in a process mediated by the protective antigen and specific cell receptors. In the cytosol, the lethal factor cleaves the N-terminal tail of many MAPK kinases, thus deranging a major cell signaling pathway. The structural features at the basis of these activities of LF are reviewed here with particular attention to the proteolytic activity and to the identification of specific inhibitors. A significant similarity between the metalloprotease domain of the lethal factor and of that of the clostridial neurotoxins has been noted and is discussed.


Subject(s)
Antigens, Bacterial/metabolism , Bacillus anthracis/enzymology , Bacterial Toxins/metabolism , Metalloproteases/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Animals , Anthrax/enzymology , Anthrax/microbiology , Antigens, Bacterial/chemistry , Bacillus anthracis/pathogenicity , Bacterial Toxins/chemistry , Binding Sites , Crystallography, X-Ray , Metalloproteases/chemistry , Mitogen-Activated Protein Kinase Kinases/chemistry , Models, Molecular , Protein Conformation
13.
J Biol Chem ; 284(38): 25761-71, 2009 Sep 18.
Article in English | MEDLINE | ID: mdl-19620708

ABSTRACT

Anthrax lethal toxin (LT) was previously shown to enhance transcriptional activity of NF-kappaB in tumor necrosis factor-alpha-activated primary human endothelial cells. Here we show that this LT-mediated increase in NF-kappaB activation is associated with the enhanced degradation of the inhibitory proteins IkappaBalpha and IkappaBbeta but not IkappaBepsilon. Moreover, this was accompanied by enhanced activation of the IkappaB kinase complex (IKK), which is responsible for targeting IkappaB proteins for degradation. Importantly, LT enhancement of IkappaBalpha degradation was completely blocked by a selective IKKbeta inhibitor, whereas IkappaBbeta degradation was attenuated, suggesting a mechanistic link. Consistent with the above data, LT-cotreated cells show elevated phosphorylation of two IKK substrates, IkappaBalpha and p65, both of which were blocked by incubation with the IKKbeta inhibitor. Consistent with NF-kappaB activation, LT increased transcription of the NF-kappaB regulated gene CD40. Conversely, LT inhibited transcription of another NF-kappaB-regulated gene, CCL2. This inhibition was linked to the LT-mediated suppression of another CCL2-regulating transcription factor, AP-1 (activator protein-1). These data suggest that LT-mediated enhancement of NF-kappaB is IKK-dependent, but importantly, the net effect of LT on the transcription of proinflammatory genes is driven by the cumulative effect of LT on the particular set of transcription factors that regulate a given promoter. Together, these findings provide new mechanistic insight on how LT may disrupt the host response to anthrax.


Subject(s)
Antigens, Bacterial/pharmacology , Bacterial Toxins/pharmacology , CD40 Antigens/biosynthesis , Chemokine CCL2/biosynthesis , Endothelial Cells/enzymology , Endothelium, Vascular/enzymology , Gene Expression Regulation/drug effects , I-kappa B Kinase/metabolism , Anthrax/enzymology , Antigens, Bacterial/metabolism , Bacterial Toxins/metabolism , Cells, Cultured , Enzyme Activation/drug effects , Humans , I-kappa B Proteins/metabolism , Inflammation/enzymology , Transcription Factor AP-1/metabolism , Transcription Factor RelA/metabolism , Transcription, Genetic/drug effects
14.
J Biol Chem ; 284(19): 12874-85, 2009 May 08.
Article in English | MEDLINE | ID: mdl-19269962

ABSTRACT

The modulation of cellular processes by small molecule inhibitors, gene inactivation, or targeted knockdown strategies combined with phenotypic screens are powerful approaches to delineate complex cellular pathways and to identify key players involved in disease pathogenesis. Using chemical genetic screening, we tested a library of known phosphatase inhibitors and identified several compounds that protected Bacillus anthracis infected macrophages from cell death. The most potent compound was assayed against a panel of sixteen different phosphatases of which CD45 was found to be most sensitive to inhibition. Testing of a known CD45 inhibitor and antisense phosphorodiamidate morpholino oligomers targeting CD45 also protected B. anthracis-infected macrophages from cell death. However, reduced CD45 expression did not protect anthrax lethal toxin (LT) treated macrophages, suggesting that the pathogen and independently added LT may signal through distinct pathways. Subsequent, in vivo studies with both gene-targeted knockdown of CD45 and genetically engineered mice expressing reduced levels of CD45 resulted in protection of mice after infection with the virulent Ames B. anthracis. Intermediate levels of CD45 expression were critical for the protection, as mice expressing normal levels of CD45 or disrupted CD45 phosphatase activity or no CD45 all succumbed to this pathogen. Mechanism-based studies suggest that the protection provided by reduced CD45 levels results from regulated immune cell homeostasis that may diminish the impact of apoptosis during the infection. To date, this is the first report demonstrating that reduced levels of host phosphatase CD45 modulate anthrax pathogenesis.


Subject(s)
Anthrax/enzymology , Anthrax/prevention & control , Bacillus anthracis/pathogenicity , Leukocyte Common Antigens/metabolism , Animals , Antigens, Bacterial/metabolism , Antigens, Bacterial/toxicity , Apoptosis , Bacillus anthracis/physiology , Bacterial Toxins/antagonists & inhibitors , Bacterial Toxins/metabolism , Bacterial Toxins/toxicity , Cell Survival , Female , Flow Cytometry , Gene Knockdown Techniques , Genetic Testing , Immunoblotting , Immunoenzyme Techniques , Leukocyte Common Antigens/antagonists & inhibitors , Macrophages/microbiology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Morpholines/pharmacology , Morpholinos , Phagocytosis , Phosphoric Monoester Hydrolases/metabolism , Spores, Bacterial/growth & development , Spores, Bacterial/pathogenicity
15.
Proc Natl Acad Sci U S A ; 105(52): 20816-21, 2008 Dec 30.
Article in English | MEDLINE | ID: mdl-19075243

ABSTRACT

Type I IFNs were discovered as the primary antiviral cytokines and are now known to serve critical functions in host defense against bacterial pathogens. Accordingly, established mediators of IFN antiviral activity may mediate previously unrecognized antibacterial functions. RNase-L is the terminal component of an RNA decay pathway that is an important mediator of IFN-induced antiviral activity. Here, we identify a role for RNase-L in the host antibacterial response. RNase-L(-/-) mice exhibited a dramatic increase in mortality after challenge with Bacillus anthracis and Escherichia coli; this increased susceptibility was due to a compromised immune response resulting in increased bacterial load. Investigation of the mechanisms of RNase-L antibacterial activity indicated that RNase-L is required for the optimal induction of proinflammatory cytokines that play essential roles in host defense from bacterial pathogens. RNase-L also regulated the expression of the endolysosomal protease, cathepsin-E, and endosome-associated activities, that function to eliminate internalized bacteria and may contribute to RNase-L antimicrobial action. Our results reveal a unique role for RNase-L in the antibacterial response that is mediated through multiple mechanisms. As a regulator of fundamental components of the innate immune response, RNase-L represents a viable therapeutic target to augment host defense against diverse microbial pathogens.


Subject(s)
Anthrax/enzymology , Bacillus anthracis , Endoribonucleases/biosynthesis , Escherichia coli Infections/enzymology , Escherichia coli , Interferon Type I/biosynthesis , Animals , Anthrax/genetics , Anthrax/immunology , Bacillus anthracis/immunology , Cathepsin E/biosynthesis , Cathepsin E/genetics , Cathepsin E/immunology , Endoribonucleases/genetics , Endoribonucleases/immunology , Endosomes/enzymology , Endosomes/genetics , Endosomes/immunology , Escherichia coli/immunology , Escherichia coli Infections/genetics , Escherichia coli Infections/immunology , Gene Expression Regulation, Enzymologic/genetics , Gene Expression Regulation, Enzymologic/immunology , Interferon Type I/genetics , Interferon Type I/immunology , Mice , Mice, Knockout , RNA Stability/genetics , RNA Stability/immunology
16.
J Microbiol Biotechnol ; 18(4): 778-83, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18467876

ABSTRACT

Anthrax is an infectious disease caused by toxigenic strains of the Gram-positive bacterium Bacillus anthracis. To identify the mitochondrial proteins that are expressed differently in murine macrophages infected with spores of B. anthracis Sterne, proteomic and MALDI-TOF/MS analyses of uninfected and infected macrophages were conducted. As a result, 13 mitochondrial proteins with different expression patterns were discovered in the infected murine macrophages, and some were identified as ATP5b, NIAP-5, ras-related GTP binding protein B isoform CRAa, along with several unnamed proteins. Among these proteins, ATP5b is related to energy production and cytoskeletal rearrangement, whereas NIAP-5 causes apoptosis of host cells due to binding with caspase-9. Therefore, this paper focused on ATP5b, which was found to be downregulated following infection. The downregulated ATP5b also reduced ATP production in the murine macrophages infected with B. anthracis spores. Consequently, this study represents the first mitochondrial proteome analysis of infected macrophages.


Subject(s)
Anthrax/microbiology , Bacillus anthracis/physiology , Macrophages/enzymology , Macrophages/microbiology , Mitochondrial Proton-Translocating ATPases/metabolism , Animals , Anthrax/enzymology , Anthrax/metabolism , Cell Line, Tumor , Electrophoresis, Gel, Two-Dimensional , Humans , Macrophages/chemistry , Macrophages/metabolism , Mice , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/metabolism , Mitochondrial Proton-Translocating ATPases/chemistry , Peptide Mapping , Proteomics , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Spores, Bacterial/physiology
17.
J Biol Chem ; 283(15): 9531-42, 2008 Apr 11.
Article in English | MEDLINE | ID: mdl-18263586

ABSTRACT

Pathology data from the anthrax animal models show evidence of significant increases in vascular permeability coincident with hemostatic imbalances manifested by thrombocytopenia, transient leucopenia, and aggressive disseminated intravascular coagulation. In this study we hypothesized that anthrax infection modulates the activity of von Willebrand factor (VWF) and its endogenous regulator ADAMTS13, which play important roles in hemostasis and thrombosis, including interaction of endothelial cells with platelets. We previously demonstrated that purified anthrax neutral metalloproteases Npr599 and InhA are capable of cleaving a variety of host structural and regulatory proteins. Incubation of human plasma with these proteases at 37 degrees C in the presence of urea as a mild denaturant results in proteolysis of VWF. Also in these conditions, InhA directly cleaves plasma ADAMTS13 protein. Npr599 and InhA digest synthetic VWF substrate FRETS-VWF73. Amino acid sequencing of VWF fragments produced by InhA suggests that one of the cleavage sites of VWF is located at domain A2, the target domain of ADAMTS13. Proteolysis of VWF by InhA impairs its collagen binding activity (VWF:CBA) and ristocetin-induced platelet aggregation activity. In plasma from anthrax spore-challenged DBA/2 mice, VWF antigen levels increase up to 2-fold at day 3 post-infection with toxigenic Sterne 34F(2) strain, whereas VWF:CBA levels drop in a time-dependent manner, suggesting dysfunction of VWF instead of its quantitative deficiency. This conclusion is further supported by significant reduction in the amount of VWF circulating in blood in the ultra-large forms. In addition, Western blot analysis shows proteolytic depletion of ADAMTS13 from plasma of spore-challenged mice despite its increased expression in the liver. Our results suggest a new mechanism of anthrax coagulopathy affecting the levels and functional activities of both VWF and its natural regulator ADAMTS13. This mechanism may contribute to hemorrhage and thrombosis typical in anthrax.


Subject(s)
ADAM Proteins/metabolism , Anthrax/enzymology , Bacterial Proteins/metabolism , Disseminated Intravascular Coagulation/enzymology , Metalloproteases/metabolism , von Willebrand Factor/metabolism , ADAMTS13 Protein , Animals , Anthrax/pathology , Anti-Bacterial Agents/pharmacology , Blood Platelets/metabolism , Blood Platelets/microbiology , Blood Platelets/pathology , Cell Communication/drug effects , Collagen/metabolism , Disease Models, Animal , Disseminated Intravascular Coagulation/microbiology , Disseminated Intravascular Coagulation/pathology , Endothelial Cells/metabolism , Endothelial Cells/microbiology , Endothelial Cells/pathology , Hemostasis/drug effects , Humans , Leukopenia/enzymology , Leukopenia/microbiology , Leukopenia/pathology , Metalloendopeptidases/metabolism , Mice , Plasma/enzymology , Plasma/microbiology , Platelet Aggregation/drug effects , Protein Binding/drug effects , Protein Structure, Tertiary , Ristocetin/pharmacology , Spores, Bacterial/enzymology , Substrate Specificity/drug effects , Thrombocytopenia/enzymology , Thrombocytopenia/microbiology , Thrombocytopenia/pathology , Thrombosis/enzymology , Thrombosis/microbiology , Thrombosis/pathology , Time Factors , Urea/pharmacology
18.
PLoS Pathog ; 3(12): e187, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18069891

ABSTRACT

Bacillus anthracis, the etiological agent of anthrax, is a spore-forming gram-positive bacterium. Infection with this pathogen results in multisystem dysfunction and death. The pathogenicity of B. anthracis is due to the production of virulence factors, including edema toxin (ET). Recently, we established the protective role of type-IIA secreted phospholipase A2 (sPLA2-IIA) against B. anthracis. A component of innate immunity produced by alveolar macrophages (AMs), sPLA2-IIA is found in human and animal bronchoalveolar lavages at sufficient levels to kill B. anthracis. However, pulmonary anthrax is almost always fatal, suggesting the potential impairment of sPLA2-IIA synthesis and/or action by B. anthracis factors. We investigated the effect of purified ET and ET-deficient B. anthracis strains on sPLA2-IIA expression in primary guinea pig AMs. We report that ET inhibits sPLA2-IIA expression in AMs at the transcriptional level via a cAMP/protein kinase A-dependent process. Moreover, we show that live B. anthracis strains expressing functional ET inhibit sPLA2-IIA expression, whereas ET-deficient strains induced this expression. This stimulatory effect, mediated partly by the cell wall peptidoglycan, can be counterbalanced by ET. We conclude that B. anthracis down-regulates sPLA2-IIA expression in AMs through a process involving ET. Our study, therefore, describes a new molecular mechanism implemented by B. anthracis to escape innate host defense. These pioneering data will provide new molecular targets for future intervention against this deadly pathogen.


Subject(s)
Anthrax/prevention & control , Antigens, Bacterial/toxicity , Bacillus anthracis/enzymology , Bacterial Toxins/toxicity , Group II Phospholipases A2/metabolism , Macrophages, Alveolar/drug effects , Animals , Anthrax/enzymology , Anthrax/immunology , Bronchoalveolar Lavage , Cells, Cultured , Down-Regulation , Gene Expression Regulation, Bacterial , Gene Silencing , Group II Phospholipases A2/immunology , Guinea Pigs , Host-Pathogen Interactions , Immunity, Innate , Macrophages, Alveolar/enzymology , Macrophages, Alveolar/microbiology , Male , RNA, Messenger/metabolism
19.
Int J Biochem Cell Biol ; 39(1): 20-4, 2007.
Article in English | MEDLINE | ID: mdl-17008119

ABSTRACT

Lethal factor (LF), along with its receptor-binding partner protective antigen (PA), forms lethal toxin (LT), a critical virulence factor for Bacillus anthracis. LF is a Zn(2+) protease that cleaves specific mitogen activated protein kinase kinases (MAPKKs), inactivating signal transduction intermediates required for normal immune function. Initial research emphasized the role of LT in attenuating pro-inflammatory responses by macrophages, the primary targets of infection. More recent studies have revealed that LT affects a broad range of immune cells. In addition to direct effects on macrophages and neutrophils, LT suppresses the costimulatory functions of dendritic cells, thereby impeding essential cross-talk between innate and adaptive immune responses. Moreover, LT acts directly on T and B lymphocytes, blocking antigen receptor-dependent proliferation, cytokine production and Ig production. In this manner, LT mounts a broad-based attack on host immunity, thus providing B. anthracis with multiple mechanisms for avoiding protective host responses.


Subject(s)
Anthrax/immunology , Antigens, Bacterial/immunology , Bacillus anthracis/immunology , Bacterial Toxins/immunology , Leukocytes, Mononuclear/immunology , MAP Kinase Signaling System/immunology , Metalloproteases/immunology , Animals , Anthrax/enzymology , Anthrax/metabolism , Antigens, Bacterial/metabolism , Bacillus anthracis/enzymology , Bacterial Toxins/metabolism , Humans , Inflammation/enzymology , Inflammation/immunology , Metalloproteases/metabolism , Mitogen-Activated Protein Kinase Kinases/immunology , Mitogen-Activated Protein Kinase Kinases/metabolism
20.
Infect Immun ; 74(7): 3756-64, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16790747

ABSTRACT

Several models of anthrax pathogenesis suggest that early in the infectious process Bacillus anthracis endospores germinate and outgrow into vegetative bacilli within phagocytes before being released into the blood. Here, we define the respective contributions of three phospholipases C (PLCs) to the pathogenesis of B. anthracis. Genetic deletions of the PLCs were made in the Sterne 7702 background, resulting in the respective loss of their activities. The PLCs were redundant both in tissue culture and in murine models of anthrax. Deletion of all three PLC genes was required for attenuation of virulence in mice after intratracheal inoculation. This attenuation may be attributed to the inability of the PLC-null strain to grow in association with the macrophage. Complementation of these defects in both models of anthrax was achieved by expression of the PLC genes in trans. The functional redundancy between PLCs in the virulence of B. anthracis implies that their activities are important for anthrax pathogenesis.


Subject(s)
Anthrax/enzymology , Anthrax/microbiology , Bacillus anthracis/enzymology , Bacillus anthracis/growth & development , Macrophages/microbiology , Type C Phospholipases/physiology , Animals , Bacillus anthracis/pathogenicity , Bone Marrow Cells/microbiology , Disease Models, Animal , Female , Gene Deletion , Intubation, Intratracheal , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Type C Phospholipases/genetics , Virulence
SELECTION OF CITATIONS
SEARCH DETAIL
...