Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
J Clin Invest ; 134(9)2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38502193

ABSTRACT

Chimeric antigen receptor (CAR) designs that incorporate pharmacologic control are desirable; however, designs suitable for clinical translation are needed. We designed a fully human, rapamycin-regulated drug product for targeting CD33+ tumors called dimerizaing agent-regulated immunoreceptor complex (DARIC33). T cell products demonstrated target-specific and rapamycin-dependent cytokine release, transcriptional responses, cytotoxicity, and in vivo antileukemic activity in the presence of as little as 1 nM rapamycin. Rapamycin withdrawal paused DARIC33-stimulated T cell effector functions, which were restored following reexposure to rapamycin, demonstrating reversible effector function control. While rapamycin-regulated DARIC33 T cells were highly sensitive to target antigen, CD34+ stem cell colony-forming capacity was not impacted. We benchmarked DARIC33 potency relative to CD19 CAR T cells to estimate a T cell dose for clinical testing. In addition, we integrated in vitro and preclinical in vivo drug concentration thresholds for off-on state transitions, as well as murine and human rapamycin pharmacokinetics, to estimate a clinically applicable rapamycin dosing schedule. A phase I DARIC33 trial has been initiated (PLAT-08, NCT05105152), with initial evidence of rapamycin-regulated T cell activation and antitumor impact. Our findings provide evidence that the DARIC platform exhibits sensitive regulation and potency needed for clinical application to other important immunotherapy targets.


Subject(s)
Leukemia, Myeloid, Acute , Sialic Acid Binding Ig-like Lectin 3 , Sirolimus , T-Lymphocytes , Animals , Female , Humans , Male , Mice , Immunotherapy, Adoptive , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/therapy , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Receptors, Chimeric Antigen/immunology , Sialic Acid Binding Ig-like Lectin 3/immunology , Sialic Acid Binding Ig-like Lectin 3/metabolism , Sirolimus/pharmacology , Sirolimus/administration & dosage , T-Lymphocytes/immunology , T-Lymphocytes/drug effects , Xenograft Model Antitumor Assays
2.
J Infect Dis ; 217(2): 270-279, 2018 01 04.
Article in English | MEDLINE | ID: mdl-29099935

ABSTRACT

Background: Necrotizing fasciitis (NF) retains a very high mortality rate despite prompt and adequate antibiotic treatment and surgical debridement. Necrotizing fasciitis has recently been associated with Streptococcus dysgalactiae subspecies equisimilis (SDSE). Methods: We investigated the causes of a very severe clinical manifestation of SDSE-NF by assessing both host and pathogen factors. Results: We found a lack of streptokinase-function blocking antibodies in the patient resulting in increased streptokinase-mediated fibrinolysis and bacterial spread. At the same time, the clinical SDSE isolate produced very high levels of streptokinase. Exogenous immunoglobulin Gs (ex-IgGs) efficiently blocked streptokinase-mediated fibrinolysis in vitro, indicating a protective role against the action of streptokinase. In vivo, SDSE infection severity was also attenuated by ex-IgGs in a NF mouse model. Conclusions: These findings illustrate for the first time that the lack of specific antibodies against streptococcal virulence factors, such as streptokinase, may contribute to NF disease severity. This can be counteracted by ex-IgGs.


Subject(s)
Antibodies, Bacterial/immunology , Fasciitis, Necrotizing/pathology , Streptococcal Infections/pathology , Streptococcus/pathogenicity , Streptokinase/antagonists & inhibitors , Virulence Factors/antagonists & inhibitors , Adult , Animals , Fasciitis, Necrotizing/microbiology , Female , Fibrinolytic Agents/immunology , Fibrinolytic Agents/metabolism , Host-Pathogen Interactions , Humans , Mice, Inbred C57BL , Streptococcal Infections/microbiology , Streptococcus/immunology , Streptokinase/immunology , Virulence Factors/immunology
3.
PLoS Pathog ; 13(9): e1006603, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28945820

ABSTRACT

Various bacterial toxins circumvent host defenses through overproduction of cAMP. In a previous study, we showed that edema factor (EF), an adenylate cyclase from Bacillus anthracis, disrupts endocytic recycling mediated by the small GTPase Rab11. As a result, cargo proteins such as cadherins fail to reach inter-cellular junctions. In the present study, we provide further mechanistic dissection of Rab11 inhibition by EF using a combination of Drosophila and mammalian systems. EF blocks Rab11 trafficking after the GTP-loading step, preventing a constitutively active form of Rab11 from delivering cargo vesicles to the plasma membrane. Both of the primary cAMP effector pathways -PKA and Epac/Rap1- contribute to inhibition of Rab11-mediated trafficking, but act at distinct steps of the delivery process. PKA acts early, preventing Rab11 from associating with its effectors Rip11 and Sec15. In contrast, Epac functions subsequently via the small GTPase Rap1 to block fusion of recycling endosomes with the plasma membrane, and appears to be the primary effector of EF toxicity in this process. Similarly, experiments conducted in mammalian systems reveal that Epac, but not PKA, mediates the activity of EF both in cell culture and in vivo. The small GTPase Arf6, which initiates endocytic retrieval of cell adhesion components, also contributes to junctional homeostasis by counteracting Rab11-dependent delivery of cargo proteins at sites of cell-cell contact. These studies have potentially significant practical implications, since chemical inhibition of either Arf6 or Epac blocks the effect of EF in cell culture and in vivo, opening new potential therapeutic avenues for treating symptoms caused by cAMP-inducing toxins or related barrier-disrupting pathologies.


Subject(s)
Antigens, Bacterial/pharmacology , Bacterial Toxins/pharmacology , Edema/metabolism , Endosomes/drug effects , Intercellular Junctions/drug effects , ADP-Ribosylation Factor 6 , ADP-Ribosylation Factors/metabolism , Adenylyl Cyclases/metabolism , Animals , Cadherins/metabolism , Cell Line , Endosomes/metabolism , Intercellular Junctions/metabolism , Protein Transport/drug effects , rab GTP-Binding Proteins/metabolism
4.
J Biol Chem ; 291(27): 13964-13973, 2016 Jul 01.
Article in English | MEDLINE | ID: mdl-27226531

ABSTRACT

Emerging antibiotic resistance among pathogenic bacteria is an issue of great clinical importance, and new approaches to therapy are urgently needed. Anacardic acid, the primary active component of cashew nut shell extract, is a natural product used in the treatment of a variety of medical conditions, including infectious abscesses. Here, we investigate the effects of this natural product on the function of human neutrophils. We find that anacardic acid stimulates the production of reactive oxygen species and neutrophil extracellular traps, two mechanisms utilized by neutrophils to kill invading bacteria. Molecular modeling and pharmacological inhibitor studies suggest anacardic acid stimulation of neutrophils occurs in a PI3K-dependent manner through activation of surface-expressed G protein-coupled sphingosine-1-phosphate receptors. Neutrophil extracellular traps produced in response to anacardic acid are bactericidal and complement select direct antimicrobial activities of the compound.


Subject(s)
Anacardic Acids/pharmacology , Anacardium/chemistry , Anti-Bacterial Agents/pharmacology , Extracellular Traps/metabolism , Neutrophils/drug effects , Humans , Lysophospholipids/metabolism , Respiratory Burst , Sphingosine/analogs & derivatives , Sphingosine/metabolism
5.
Nat Commun ; 6: 8369, 2015 Oct 13.
Article in English | MEDLINE | ID: mdl-26458291

ABSTRACT

Tamoxifen is a selective oestrogen receptor modulator widely used for the treatment of breast cancer. In addition to its activity as an oestrogen receptor agonist/antagonist, tamoxifen also modulates sphingolipid biosynthesis, which has been shown to play an important role in the regulation of neutrophil activity. Here, we find that tamoxifen stimulation enhances several pro-inflammatory pathways in human neutrophils, including chemotaxis, phagocytosis and neutrophil extracellular trap (NET) formation. The enhancement of NET production occurs via a ceramide/PKCζ-mediated pathway, and treatment with synthetic ceramide is sufficient to promote NET formation. Pretreatment of human neutrophils with tamoxifen boosts neutrophil bactericidal capacity against a variety of pathogens in vitro and enhances clearance of the leading human pathogen methicillin-resistant Staphylococcus aureus in vivo. Our results suggest that tamoxifen, and the lipid signalling pathways it modulates, merit further exploration as targets for boosting host innate immune function.


Subject(s)
Ceramides/metabolism , Extracellular Traps/drug effects , Neutrophils/drug effects , Selective Estrogen Receptor Modulators/pharmacology , Tamoxifen/pharmacology , Animals , Female , Healthy Volunteers , Humans , Immunity, Innate/drug effects , Methicillin-Resistant Staphylococcus aureus , Mice , Neutrophils/metabolism , Protein Kinase C/metabolism
6.
EBioMedicine ; 2(7): 690-8, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26288841

ABSTRACT

Antibiotic resistance poses an increasingly grave threat to the public health. Of pressing concern, rapid spread of carbapenem-resistance among multidrug-resistant (MDR) Gram-negative rods (GNR) is associated with few treatment options and high mortality rates. Current antibiotic susceptibility testing guiding patient management is performed in a standardized manner, identifying minimum inhibitory concentrations (MIC) in bacteriologic media, but ignoring host immune factors. Lacking activity in standard MIC testing, azithromycin (AZM), the most commonly prescribed antibiotic in the U.S., is never recommended for MDR GNR infection. Here we report a potent bactericidal action of AZM against MDR carbapenem-resistant isolates of Pseudomonas aeruginosa, Klebsiella pneumoniae, and Acinetobacter baumannii. This pharmaceutical activity is associated with enhanced AZM cell penetration in eukaryotic tissue culture media and striking multi-log-fold synergies with host cathelicidin antimicrobial peptide LL-37 or the last line antibiotic colistin. Finally, AZM monotherapy exerts clear therapeutic effects in murine models of MDR GNR infection. Our results suggest that AZM, currently ignored as a treatment option, could benefit patients with MDR GNR infections, especially in combination with colistin.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antimicrobial Cationic Peptides/pharmacology , Azithromycin/pharmacology , Drug Resistance, Multiple, Bacterial/drug effects , Gram-Negative Bacteria/drug effects , Animals , Antimicrobial Cationic Peptides/therapeutic use , Azithromycin/therapeutic use , Cathelicidins/pharmacology , Cathelicidins/therapeutic use , Cell Membrane Permeability/drug effects , Colistin/pharmacology , Colistin/therapeutic use , Culture Media , Disease Models, Animal , Drug Synergism , Female , Gram-Negative Bacteria/ultrastructure , Humans , Mice, Inbred C57BL , Microbial Sensitivity Tests
7.
mBio ; 6(2): e00133, 2015 Mar 10.
Article in English | MEDLINE | ID: mdl-25759502

ABSTRACT

UNLABELLED: The M1T1 clone of group A Streptococcus (GAS) is associated with severe invasive infections, including necrotizing fasciitis and septicemia. During invasive M1T1 GAS disease, mutations in the covRS regulatory system led to upregulation of an ADP-ribosyltransferase, SpyA. Surprisingly, a GAS ΔspyA mutant was resistant to killing by macrophages and caused higher mortality with impaired bacterial clearance in a mouse intravenous challenge model. GAS expression of SpyA triggered macrophage cell death in association with caspase-1-dependent interleukin 1ß (IL-1ß) production, and differences between wild-type (WT) and ΔspyA GAS macrophage survival levels were lost in cells lacking caspase-1, NOD-like receptor protein 3 (NLRP3), apoptosis-associated speck-like protein (ASC), or pro-IL-1ß. Similar in vitro findings were identified in macrophage studies performed with pseudomonal exotoxin A, another ADP-ribosylating toxin. Thus, SpyA triggers caspase-1-dependent inflammatory cell death in macrophages, revealing a toxin-triggered IL-1ß-dependent innate immune response pathway critical in defense against invasive bacterial infection. IMPORTANCE: Group A Streptococcus (GAS) is a leading human pathogen capable of producing invasive infections even in healthy individuals. GAS bacteria produce a toxin called SpyA that modifies host proteins through a process called ADP ribosylation. We describe how macrophages, frontline defenders of the host innate immune system, respond to SpyA by undergoing a specialized form of cell death in which they are activated to release the proinflammatory cytokine molecule interleukin 1ß (IL-1ß). Release of IL-1ß activates host immune cell clearance of GAS, as we demonstrated in tissue culture models of macrophage bacterial killing and in vivo mouse infectious-challenge experiments. Similar macrophage responses to a related toxin of Pseudomonas bacteria were also shown. Thus, macrophages recognize certain bacterial toxins to activate a protective immune response in the host.


Subject(s)
ADP Ribose Transferases/immunology , Interleukin-1beta/metabolism , Macrophages/immunology , Macrophages/microbiology , Streptococcus pyogenes/enzymology , Streptococcus pyogenes/immunology , ADP Ribose Transferases/genetics , Animals , Cell Survival , Disease Models, Animal , Gene Deletion , Mice , Streptococcal Infections/immunology , Streptococcal Infections/microbiology , Streptococcus pyogenes/genetics , Virulence
8.
FASEB J ; 29(5): 1859-68, 2015 May.
Article in English | MEDLINE | ID: mdl-25630970

ABSTRACT

Bicuspid aortic valves calcify at a significantly higher rate than normal aortic valves, a process that involves increased inflammation. Because we have previously found that bicuspid aortic valve experience greater stretch, we investigated the potential connection between stretch and inflammation in human aortic valve interstitial cells (AVICs). Microarray, quantitative PCR (qPCR), and protein assays performed on AVICs exposed to cyclic stretch showed that stretch was sufficient to increase expression of interleukin and metalloproteinase family members by more than 1.5-fold. Conditioned medium from stretched AVICs was sufficient to activate leukocytes. microRNA sequencing and qPCR experiments demonstrated that miR-148a-3p was repressed in both stretched AVICs (43% repression) and, as a clinical correlate, human bicuspid aortic valves (63% reduction). miR-148a-3p was found to be a novel repressor of IKBKB based on data from qPCR, luciferase, and Western blot experiments. Furthermore, increasing miR-148a-3p levels in AVICs was sufficient to decrease NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells) signaling and NF-κB target gene expression. Our data demonstrate that stretch-mediated activation of inflammatory pathways is at least partly the result of stretch-repression of miR-148a-3p and a consequent failure to repress IKBKB. To our knowledge, we are the first to report that cyclic stretch of human AVICs activates inflammatory genes in a tissue-autonomous manner via a microRNA that regulates a central inflammatory pathway.


Subject(s)
Aortic Valve/abnormalities , Biomarkers/metabolism , Heart Valve Diseases/metabolism , I-kappa B Kinase/metabolism , Inflammation/genetics , MicroRNAs/genetics , NF-kappa B/metabolism , Aortic Valve/immunology , Aortic Valve/metabolism , Bicuspid Aortic Valve Disease , Blotting, Western , Cell Differentiation , Cell Proliferation , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Gene Expression Profiling , Heart Valve Diseases/immunology , Humans , I-kappa B Kinase/genetics , Inflammation/immunology , Inflammation/pathology , Monocytes/cytology , Monocytes/metabolism , NF-kappa B/genetics , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Stress, Physiological
9.
Infect Immun ; 82(3): 1132-40, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24366250

ABSTRACT

Bacillus anthracis, the causative agent of anthrax, relies on multiple virulence factors to subvert the host immune defense. Using Caenorhabditis elegans as an infection model, we screened approximately 5,000 transposon mutants of B. anthracis Sterne for decreased virulence. One of the attenuated mutants resulted in loss of expression of yceG and yceH, the last two genes in a six-gene cluster of tellurite resistance genes. We generated an analogous insertional mutant to confirm the phenotype and characterize the role of yceGH in resistance to host defenses. Loss of yceGH rendered the mutants more sensitive to tellurite toxicity as well as to host defenses such as reactive oxygen species and the cathelicidin family of antimicrobial peptides. Additionally, we see decreased survival in mammalian models of infection, including human whole blood and in mice. We identify a novel role for the yceGH genes in B. anthracis Sterne virulence and suggest that C. elegans is a useful infection model to study anthrax pathogenesis.


Subject(s)
Anthrax/immunology , Bacillus anthracis/genetics , Bacillus anthracis/immunology , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Tellurium/immunology , Animals , Anthrax/microbiology , Caenorhabditis elegans/immunology , Caenorhabditis elegans/microbiology , Immunity, Innate/genetics , Immunity, Innate/immunology , Mutation/genetics , Mutation/immunology , Virulence/genetics , Virulence/immunology , Virulence Factors/genetics , Virulence Factors/immunology
10.
J Inflamm (Lond) ; 10(1): 28, 2013 Aug 10.
Article in English | MEDLINE | ID: mdl-23937964

ABSTRACT

BACKGROUND: Hypoxia inducible factor-1 (HIF-1) is a major regulator of the cellular adaption to low oxygen stress and the innate immune function of myeloid cells. Treatment with the novel HIF-1 stabilizing drug AKB-4924 has been shown to enhance the bactericidal activity of keratinocytes as well as phagocytic cells. In this study, we sought to investigate the effect of pharmacological boosting of HIF-1 with AKB-4924 in keratinocytes and their contribution to the innate immune response. FINDINGS: Treatment with the novel HIF-1 stabilizing drug AKB-4924 can increase keratinocyte production of pro-inflammatory cytokines in vitro and enhance neutrophil recruitment in vivo. CONCLUSIONS: HIF plays an important role in cytokine production by keratinocytes and in neutrophil recruitment to the skin. The HIF-boosting drug AKB-4924 has the potential to enhance the immune response even in the complex environment of bacterial skin infections.

11.
Biochem J ; 455(1): 107-18, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23865566

ABSTRACT

Many bacteria have evolved ways to interact with glycosylation functions of the immune system of their hosts. Streptococcus pyogenes [GAS (group A Streptococcus)] secretes the enzyme EndoS that cleaves glycans on human IgG and impairs the effector functions of the antibody. The ndoS gene, encoding EndoS, has, until now, been thought to be conserved throughout the serotypes. However, in the present study, we identify EndoS2, an endoglycosidase in serotype M49 GAS strains. We characterized EndoS2 and the corresponding ndoS2 gene using sequencing, bioinformatics, phylogenetic analysis, recombinant expression and LC-MS analysis of glycosidic activity. This revealed that EndoS2 is present exclusively, and highly conserved, in serotype M49 of GAS and is only 37% identical with EndoS. EndoS2 showed endo-ß-N-acetylglucosaminidase activity on all N-linked glycans of IgG and on biantennary and sialylated glycans of AGP (α1-acid glycoprotein). The enzyme was found to act only on native IgG and AGP and to be specific for free biantennary glycans with or without terminal sialylation. GAS M49 expression of EndoS2 was monitored in relation to carbohydrates present in the culture medium and was linked to the presence of sucrose. We conclude that EndoS2 is a unique endoglycosidase in serotype M49 and differs from EndoS of other GAS strains by targeting both IgG and AGP. EndoS2 expands the repertoire of GAS effectors that modify key glycosylated molecules of host defence.


Subject(s)
Bacterial Proteins/metabolism , Glycoside Hydrolases/metabolism , Immunoglobulin G/metabolism , Orosomucoid/metabolism , Streptococcus pyogenes/enzymology , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Carbohydrate Sequence , Conserved Sequence , Escherichia coli/genetics , Escherichia coli/metabolism , Glycoside Hydrolases/chemistry , Glycoside Hydrolases/genetics , Host-Pathogen Interactions , Humans , Immunoglobulin G/chemistry , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Molecular Sequence Data , Orosomucoid/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Streptococcus pyogenes/chemistry , Streptococcus pyogenes/genetics , Substrate Specificity , Sucrose/metabolism
12.
PLoS Pathog ; 9(7): e1003469, 2013.
Article in English | MEDLINE | ID: mdl-23853591

ABSTRACT

Recruitment of the serine protease plasmin is central to the pathogenesis of many bacterial species, including Group A streptococcus (GAS), a leading cause of morbidity and mortality globally. A key process in invasive GAS disease is the ability to accumulate plasmin at the cell surface, however the role of host activators of plasminogen in this process is poorly understood. Here, we demonstrate for the first time that the urokinase-type plasminogen activator (uPA) contributes to plasmin recruitment and subsequent invasive disease initiation in vivo. In the absence of a source of host plasminogen activators, streptokinase (Ska) was required to facilitate cell surface plasmin acquisition by GAS. However, in the absence of Ska, host activators were sufficient to promote cell surface plasmin acquisition by GAS strain 5448 during incubation with plasminogen or human plasma. Furthermore, GAS were able mediate a significant increase in the activation of zymogen pro-uPA in human plasma. In order to assess the contribution of uPA to invasive GAS disease, a previously undescribed transgenic mouse model of infection was employed. Both C57/black 6J, and AlbPLG1 mice expressing the human plasminogen transgene, were significantly more susceptible to invasive GAS disease than uPA-/- mice. The observed decrease in virulence in uPA-/-mice was found to correlate directly with a decrease in bacterial dissemination and reduced cell surface plasmin accumulation by GAS. These findings have significant implications for our understanding of GAS pathogenesis, and research aimed at therapeutic targeting of plasminogen activation in invasive bacterial infections.


Subject(s)
Disease Resistance , Plasminogen/metabolism , Streptococcal Infections/microbiology , Streptococcus pyogenes/metabolism , Urokinase-Type Plasminogen Activator/metabolism , Animals , Crosses, Genetic , Disease Susceptibility , Enzyme Precursors/blood , Enzyme Precursors/metabolism , Fibrinolysin/metabolism , Heterozygote , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Plasminogen/genetics , Proteolysis , Streptococcal Infections/blood , Streptococcal Infections/metabolism , Streptococcus pyogenes/pathogenicity , Streptokinase/metabolism , Surface Properties , Urokinase-Type Plasminogen Activator/blood , Urokinase-Type Plasminogen Activator/genetics , Virulence
13.
FASEB J ; 27(7): 2633-43, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23531597

ABSTRACT

In Western countries, invasive infections caused by M1T1 serotype group A Streptococcus (GAS) are epidemiologically linked to mutations in the control of virulence regulatory 2-component operon (covRS). In indigenous communities and developing countries, severe GAS disease is associated with genetically diverse non-M1T1 GAS serotypes. Hypervirulent M1T1 covRS mutant strains arise through selection by human polymorphonuclear cells for increased expression of GAS virulence factors such as the DNase Sda1, which promotes neutrophil resistance. The GAS bacteremia isolate NS88.2 (emm 98.1) is a covS mutant that exhibits a hypervirulent phenotype and neutrophil resistance yet lacks the phage-encoded Sda1. Here, we have employed a comprehensive systems biology (genomic, transcriptomic, and proteomic) approach to identify NS88.2 virulence determinants that enhance neutrophil resistance in the non-M1T1 GAS genetic background. Using this approach, we have identified streptococcal collagen-like protein A and general stress protein 24 proteins as NS88.2 determinants that contribute to survival in whole blood and neutrophil resistance in non-M1T1 GAS. This study has revealed new factors that contribute to GAS pathogenicity that may play important roles in resisting innate immune defenses and the development of human invasive infections.


Subject(s)
Bacterial Proteins/immunology , Streptococcal Infections/immunology , Streptococcus pyogenes/immunology , Animals , Bacterial Adhesion/genetics , Bacterial Adhesion/immunology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Line , Electrophoresis, Gel, Two-Dimensional , Gene Expression Profiling , Gene Expression Regulation, Bacterial , Genome, Bacterial/genetics , Genomics/methods , Host-Pathogen Interactions/immunology , Humans , Mice , Microbial Viability/genetics , Microbial Viability/immunology , Mutation , Neutrophil Activation/immunology , Neutrophils/immunology , Neutrophils/metabolism , Neutrophils/microbiology , Oligonucleotide Array Sequence Analysis , Proteome/genetics , Proteome/metabolism , Proteomics/methods , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Streptococcal Infections/blood , Streptococcal Infections/microbiology , Streptococcus pyogenes/genetics , Streptococcus pyogenes/pathogenicity , Virulence/genetics , Virulence/immunology
14.
J Biol Chem ; 287(49): 40891-7, 2012 Nov 30.
Article in English | MEDLINE | ID: mdl-23038245

ABSTRACT

The bacterial pathogen Group A Streptococcus (GAS) colonizes epithelial and mucosal surfaces and can cause a broad spectrum of human disease. Through the secreted plasminogen activator streptokinase (Ska), GAS activates human plasminogen into plasmin and binds it to the bacterial surface. The resulting surface plasmin protease activity has been proposed to play a role in disrupting tissue barriers, promoting invasive spread of the bacterium. We investigated whether this surface protease activity could aid the immune evasion role through degradation of the key innate antimicrobial peptide LL-37, the human cathelicidin. Cleavage products of plasmin-degraded LL-37 were analyzed by matrix-assisted laser desorption ionization mass spectrometry. Ska-deficient GAS strains were generated by targeted allelic exchange mutagenesis and confirmed to lack surface plasmin activity after growth in human plasma or media supplemented with plasminogen and fibrinogen. Loss of surface plasmin activity left GAS unable to efficiently degrade LL-37 and increased bacterial susceptibility to killing by the antimicrobial peptide. When mice infected with GAS were simultaneously treated with the plasmin inhibitor aprotinin, a significant reduction in the size of necrotic skin lesions was observed. Together these data reveal a novel immune evasion strategy of the human pathogen: co-opting the activity of a host protease to evade peptide-based innate host defenses.


Subject(s)
Cathelicidins/chemistry , Cathelicidins/metabolism , Alleles , Amino Acid Sequence , Animals , Anticoagulants/chemistry , Antimicrobial Cationic Peptides , Fibrinolysin/chemistry , Humans , Immune System , Immunity, Innate , Kinetics , Mice , Microbial Sensitivity Tests , Molecular Sequence Data , Mutagenesis , Plasminogen/chemistry , Sequence Homology, Amino Acid , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Streptococcus pyogenes/metabolism , Streptokinase/chemistry , Streptokinase/metabolism , Virulence Factors/metabolism
15.
FASEB J ; 26(11): 4675-84, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22878963

ABSTRACT

The past 50 years has witnessed the emergence of new viral and bacterial pathogens with global effect on human health. The hyperinvasive group A Streptococcus (GAS) M1T1 clone, first detected in the mid-1980s in the United States, has since disseminated worldwide and remains a major cause of severe invasive human infections. Although much is understood regarding the capacity of this pathogen to cause disease, much less is known of the precise evolutionary events selecting for its emergence. We used high-throughput technologies to sequence a World Health Organization strain collection of serotype M1 GAS and reconstructed its phylogeny based on the analysis of core genome single-nucleotide polymorphisms. We demonstrate that acquisition of a 36-kb genome segment from serotype M12 GAS and the bacteriophage-encoded DNase Sda1 led to increased virulence of the M1T1 precursor and occurred relatively early in the molecular evolutionary history of this strain. The more recent acquisition of the phage-encoded superantigen SpeA is likely to have provided selection advantage for the global dissemination of the M1T1 clone. This study provides an exemplar for the evolution and emergence of virulent clones from microbial populations existing commensally or causing only superficial infection.


Subject(s)
Biological Evolution , Pandemics , Streptococcal Infections/epidemiology , Streptococcal Infections/microbiology , Streptococcus pyogenes/metabolism , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Line , Disease Models, Animal , Epithelial Cells/microbiology , Exotoxins/genetics , Exotoxins/metabolism , Gene Expression Regulation, Bacterial/physiology , Genome, Bacterial , Global Health , Host-Pathogen Interactions , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Neutrophils/physiology , Oligonucleotide Array Sequence Analysis , Phagocytosis , Phylogeny , Streptococcus pyogenes/classification , Streptococcus pyogenes/genetics , Streptococcus pyogenes/pathogenicity , Transcriptome , Virulence
16.
J Biol Chem ; 287(17): 13889-98, 2012 Apr 20.
Article in English | MEDLINE | ID: mdl-22371493

ABSTRACT

Staphylococcus aureus causes a wide range of human disease ranging from localized skin and soft tissue infections to potentially lethal systemic infections. S. aureus has the biosynthetic ability to generate numerous virulence factors that assist in circumventing the innate immune system during disease pathogenesis. Recent studies have uncovered a set of extracellular peptides produced by community-associated methicillin-resistant S. aureus (CA-MRSA) with homology to the phenol-soluble modulins (PSMs) from Staphylococcus epidermidis. CA-MRSA PSMs contribute to skin infection and recruit and lyse neutrophils, and truncated versions of these peptides possess antimicrobial activity. In this study, novel CA-MRSA PSM derivatives were discovered by the use of microbial imaging mass spectrometry. The novel PSM derivatives are compared with their parent full-length peptides for changes in hemolytic, cytolytic, and neutrophil-stimulating activity. A potential contribution of the major S. aureus secreted protease aureolysin in processing PSMs is demonstrated. Finally, we show that PSM processing occurs in multiple CA-MRSA strains by structural confirmation of additional novel derivatives. This work demonstrates that IMS can serve as a useful tool to go beyond genome predictions and expand our understanding of the important family of small peptide virulence factors.


Subject(s)
Mass Spectrometry/methods , Methicillin-Resistant Staphylococcus aureus/metabolism , Phenol/chemistry , Amino Acid Sequence , Animals , Anti-Infective Agents/pharmacology , Bacterial Infections/metabolism , Bacterial Proteins/chemistry , Bacterial Toxins/chemistry , Erythrocytes/cytology , Hemolysis , Humans , Immunosuppressive Agents/pharmacology , Metalloendopeptidases/chemistry , Mice , Molecular Sequence Data , Neutrophils/cytology , Neutrophils/metabolism , Sequence Homology, Amino Acid , Sheep , Skin/metabolism , Skin/microbiology , Staphylococcal Skin Infections/microbiology , Virulence Factors/chemistry
17.
J Mol Med (Berl) ; 90(9): 1079-89, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22371073

ABSTRACT

Hypoxia inducible factor-1 (HIF-1) is a transcription factor that is a major regulator of energy homeostasis and cellular adaptation to low oxygen stress. HIF-1 is also activated in response to bacterial pathogens and supports the innate immune response of both phagocytes and keratinocytes. In this work, we show that a new pharmacological compound AKB-4924 increases HIF-1 levels and enhances the antibacterial activity of phagocytes and keratinocytes against both methicillin-sensitive and methicillin-resistant strains of Staphylococcus aureus in vitro. AKB-4924 is also effective in stimulating the killing capacity of keratinocytes against the important opportunistic skin pathogens Pseudomonas aeruginosa and Acinetobacter baumanii. The effect of AKB-4924 is mediated through the activity of host cells, as the compound exerts no direct antimicrobial activity. Administered locally as a single agent, AKB-4924 limits S. aureus proliferation and lesion formation in a mouse skin abscess model. This approach to pharmacologically boost the innate immune response via HIF-1 stabilization may serve as a useful adjunctive treatment for antibiotic-resistant bacterial infections.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Hypoxia-Inducible Factor 1/immunology , Immunity, Innate/drug effects , Piperazines/therapeutic use , Pyridones/therapeutic use , Skin Diseases, Bacterial/prevention & control , Skin/microbiology , Animals , Cell Line , Female , Humans , Hypoxia-Inducible Factor 1/agonists , Keratinocytes/drug effects , Keratinocytes/immunology , Keratinocytes/microbiology , Mice , Phagocytes/drug effects , Phagocytes/immunology , Phagocytes/microbiology , Piperazines/pharmacology , Pyridones/pharmacology , Skin/drug effects , Skin/immunology , Skin Diseases, Bacterial/immunology , Staphylococcal Infections/immunology , Staphylococcal Infections/prevention & control , Staphylococcus aureus/drug effects , Staphylococcus aureus/immunology
18.
Microbiology (Reading) ; 157(Pt 9): 2485-2492, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21719540

ABSTRACT

Microbial competition exists in the general environment, such as soil or aquatic habitats, upon or within unicellular or multicellular eukaryotic life forms. The molecular actions that govern microbial competition, leading to niche establishment and microbial monopolization, remain undetermined. The emerging technology of imaging mass spectrometry (IMS) enabled the observation that there is directionality in the metabolic output of the organism Bacillus subtilis when co-cultured with Staphylococcus aureus. The directionally released antibiotic alters S. aureus virulence factor production and colonization. Therefore, IMS provides insight into the largely hidden nature of competitive microbial encounters and niche establishment, and provides a paradigm for future antibiotic discovery.


Subject(s)
Bacillus subtilis/metabolism , Microbial Viability , Staphylococcus aureus/metabolism , Animals , Bacillus subtilis/isolation & purification , Coculture Techniques , Fatty Acids/metabolism , Female , Humans , Lipopeptides/metabolism , Male , Mass Spectrometry , Mice , Microscopy, Fluorescence , Oligopeptides/metabolism , Peptides, Cyclic/metabolism , Staphylococcus aureus/isolation & purification
19.
BMC Microbiol ; 11: 120, 2011 May 27.
Article in English | MEDLINE | ID: mdl-21619648

ABSTRACT

BACKGROUND: The secreted enzyme EndoS, an endoglycosidase from Streptococcus pyogenes, hydrolyzes the N-linked glycan of the constant region of immunoglobulin G (IgG) heavy chain and renders the antibody unable to interact with Fc receptors and elicit effector functions. In this study we couple targeted allelic replacement mutagenesis and heterologous expression to elucidate the contribution of EndoS to group A Streptococcus (GAS) phagocyte resistance and pathogenicity in vitro and in vivo. RESULTS: Knocking out the EndoS gene in GAS M1T1 background revealed no significant differences in bacterial survival in immune cell killing assays or in a systemic mouse model of infection. However, exogenous addition and heterologous expression of EndoS was found to increase GAS resistance to killing by neutrophils and monocytes in vitro. Additionally, heterologous expression of EndoS in M49 GAS increased mouse virulence in vivo. CONCLUSIONS: We conclude that in a highly virulent M1T1 background, EndoS has no significant impact on GAS phagocyte resistance and pathogenicity. However, local accumulation or high levels of expression of EndoS in certain GAS strains may contribute to virulence.


Subject(s)
Glycoside Hydrolases/metabolism , Immunoglobulin G/metabolism , Phagocytes/immunology , Phagocytes/microbiology , Streptococcus pyogenes/enzymology , Streptococcus pyogenes/pathogenicity , Animals , Disease Models, Animal , Gene Deletion , Glycoside Hydrolases/genetics , Humans , Mice , Microbial Viability , Phagocytosis , Streptococcal Infections/microbiology , Streptococcal Infections/pathology , Streptococcus pyogenes/immunology , Virulence
20.
mBio ; 1(4)2010 Aug 31.
Article in English | MEDLINE | ID: mdl-20827373

ABSTRACT

The initiation of hyperinvasive disease in group A Streptococcus (GAS) serotype M1T1 occurs by mutation within the covRS two-component regulon (named covRS for control of virulence regulatory sensor kinase), which promotes resistance to neutrophil-mediated killing through the upregulation of bacteriophage-encoded Sda1 DNase. To determine whether other virulence factors contribute to this phase-switching phenomenon, we studied a panel of 10 isogenic GAS serotype M1T1 virulence gene knockout mutants. While loss of several individual virulence factors did not prevent GAS covRS switching in vivo, we found that M1 protein and hyaluronic acid capsule are indispensable for the switching phenotype, a phenomenon previously attributed uniquely to the Sda1 DNase. We demonstrate that like M1 protein and Sda1, capsule expression enhances survival of GAS serotype M1T1 within neutrophil extracellular traps. Furthermore, capsule shares with M1 protein a role in GAS resistance to human cathelicidin antimicrobial peptide LL-37. We conclude that a quorum of GAS serotype M1T1 virulence genes with cooperative roles in resistance to neutrophil extracellular killing is essential for the switch to a hyperinvasive phenotype in vivo.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Capsules/immunology , Bacterial Outer Membrane Proteins/immunology , Bacterial Proteins/genetics , Carrier Proteins/immunology , Hyaluronic Acid/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Mutation , Repressor Proteins/genetics , Streptococcal Infections/microbiology , Streptococcus pyogenes/pathogenicity , Animals , Antigens, Bacterial/genetics , Bacterial Capsules/chemistry , Bacterial Outer Membrane Proteins/genetics , Bacterial Proteins/immunology , Carrier Proteins/genetics , Female , Histidine Kinase , Humans , Intracellular Signaling Peptides and Proteins/immunology , Mice , Mice, Inbred C57BL , Microbial Viability , Neutrophils/immunology , Neutrophils/microbiology , Repressor Proteins/immunology , Streptococcal Infections/immunology , Streptococcus pyogenes/chemistry , Streptococcus pyogenes/genetics , Streptococcus pyogenes/immunology , Virulence Factors/genetics , Virulence Factors/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...