Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
1.
Methods Mol Biol ; 1124: 19-37, 2014.
Article in English | MEDLINE | ID: mdl-24504944

ABSTRACT

The development of new advances in the understanding of neutrophil biochemistry requires effective procedures for isolating purified neutrophil populations. Although methods for human neutrophil isolation are now standard, similar procedures for isolating neutrophils from many of the nonhuman species used to model human diseases are not as well developed. Since neutrophils are reactive cells, the method of isolation is extremely important to avoid isolation technique-induced alterations in cell function. We present methods here for reproducibly isolating highly purified neutrophils from large animals (bovine, equine, ovine), small animals (murine and rabbit), and nonhuman primates (cynomolgus macaques), and describe optimized details for obtaining the highest cell purity, yield, and viability. We also describe methods to verify phagocytic capacity in the purified cell populations using a flow cytometry-based phagocytosis assay.


Subject(s)
Cell Separation/methods , Neutrophils/cytology , Animals , Cattle , Horses , Humans , Immunomagnetic Separation/methods , Macaca fascicularis , Mice , Neutrophils/immunology , Rabbits , Sheep
2.
J Infect Dis ; 208(3): 520-7, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23599317

ABSTRACT

Staphylococcus aureus clonal complex 75 (herein referred to as S. argenteus) lacks the carotenoid pigment operon, crtOPQMN, responsible for production of the putative virulence factor, staphyloxanthin. Although a common cause of community-onset skin infections among Indigenous populations in northern Australia, this clone is infrequently isolated from hospital-based patients with either bacteremic or nonbacteremic infections. We hypothesized that S. argenteus would have attenuated virulence compared to other S. aureus strains due to its staphyloxanthin "deficiency." Compared to prototypical S. aureus strains, S. argenteus was more susceptible to oxidative stress and neutrophil killing in vitro and had reduced virulence in murine sepsis and skin infection models. Transformation with pTX-crtOPQMN resulted in staphyloxanthin expression and increased resistance to oxidative stress in vitro. However, neither resistance to neutrophil killing nor in vivo virulence was increased. Thus, reduced virulence of S. argenteus in these models is due to mechanisms unrelated to lack of staphyloxanthin production.


Subject(s)
Sepsis/pathology , Staphylococcal Infections/pathology , Staphylococcal Skin Infections/pathology , Staphylococcus aureus/metabolism , Staphylococcus aureus/pathogenicity , Virulence Factors/metabolism , Xanthophylls/metabolism , Animals , Australia , Child , Disease Models, Animal , Genetic Complementation Test , Humans , Male , Mice , Mice, Inbred BALB C , Operon , Sepsis/microbiology , Staphylococcal Infections/microbiology , Staphylococcal Skin Infections/microbiology , Staphylococcus aureus/genetics , Staphylococcus aureus/isolation & purification , Virulence , Virulence Factors/deficiency , Virulence Factors/genetics , Xanthophylls/deficiency , Xanthophylls/genetics
3.
J Innate Immun ; 5(2): 124-36, 2013.
Article in English | MEDLINE | ID: mdl-22986450

ABSTRACT

We demonstrated recently that Francisella tularensis profoundly impairs human neutrophil apoptosis, but how this is achieved is largely unknown. Herein we used human oligonucleotide microarrays to test the hypothesis that changes in neutrophil gene expression contribute to this phenotype, and now demonstrate that F. tularensis live vaccine strain (LVS) caused significant changes in neutrophil gene expression over a 24-hour time period relative to the uninfected controls. Of approximately 47,000 genes analyzed, 3,435 were significantly up- or downregulated by LVS, including 365 unique genes associated with apoptosis and cell survival. Specific targets in this category included genes asso-ciated with the intrinsic and extrinsic apoptotic pathways (CFLAR, TNFAIP3, TNFRSF10D, SOD2, BCL2A1, BIRC4, PIM2, TNFSF10, TNFRSF10C, CASP2 and CASP8) and genes that act via the NFĸB pathway and other mechanisms to prolong cell viability (NFKB1, NFKB2 and RELA, IL1B, CAST, CDK2,GADD45B, BCL3, BIRC3, CDK2, IL1A, PBEF1, IL6, CXCL1, CCL4 and VEGF). The microarray data were confirmed by qPCR and pathway analysis. Moreover, we demonstrate that the X-linked inhibitor of apoptosis protein remained abundant in polymorphonuclear leukocytes over 48 h of LVS infection, whereas BAX mRNA and protein were progressively downregulated. These data strongly suggest that antiapoptotic and prosurvival mechanisms collaborate to sustain the viability of F. tularensis--infected neutrophils.


Subject(s)
Francisella tularensis/physiology , Neutrophils/immunology , Tularemia/immunology , Apoptosis , Apoptosis Regulatory Proteins/genetics , Cell Survival , Cells, Cultured , Computational Biology , Gene Expression Regulation , Humans , Microarray Analysis , NF-kappa B/metabolism , Neutrophils/microbiology , Signal Transduction/genetics , Signal Transduction/immunology , Tularemia/genetics , X-Linked Inhibitor of Apoptosis Protein/genetics , X-Linked Inhibitor of Apoptosis Protein/metabolism , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
4.
J Infect Dis ; 206(8): 1185-93, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22872735

ABSTRACT

BACKGROUND: Staphylococcus aureus produces numerous molecules that facilitate survival in the host. We recently identified a novel S. aureus leukotoxin (leukotoxin GH [LukGH]) using proteomics, but its role in virulence remains unclear. Here we investigated the role of LukGH in vivo. METHODS: We tested cytotoxicity of LukGH toward polymorphonuclear leukocytes (PMNs) from mice, rabbits, monkeys, and humans. LukGH was administered to mice, rabbits, and a cynomolgus monkey by subcutaneous or intradermal injection to assess cytotoxicity or host response in vivo. The effects of LukGH in vivo were compared with those of Panton-Valentine leukocidin (PVL), a well-characterized S. aureus leukotoxin. The contribution of LukGH to S. aureus infection was tested using mouse and rabbit infection models. RESULTS: Susceptibility of PMNs to LukGH was similar between humans and cynomolgus monkeys, and was greater than that of rabbits, which in turn was greater than that of mice. LukGH or PVL caused skin inflammation in rabbits and a monkey, but deletion of neither lukGH nor lukGH and lukS/F-PV reduced severity of USA300 infections in rabbits or mice. Rather, some disease parameters (eg, rabbit abscess size) were increased following infection with a lukGH and lukS/F-PV deletion strain. CONCLUSIONS: Our findings indicate that S. aureus leukotoxins enhance the host inflammatory response and influence the outcome of infection.


Subject(s)
Exotoxins/toxicity , Inflammation/chemically induced , Staphylococcus aureus/pathogenicity , Virulence Factors/toxicity , Animals , Disease Models, Animal , Exotoxins/administration & dosage , Humans , Inflammation/immunology , Injections, Intradermal , Injections, Subcutaneous , Macaca fascicularis , Male , Mice , Neutrophils/immunology , Neutrophils/microbiology , Rabbits , Staphylococcal Infections/pathology , Virulence Factors/administration & dosage
5.
J Leukoc Biol ; 92(2): 361-74, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22581932

ABSTRACT

CA-MRSA infections are often caused by strains encoding PVL, which can cause lysis of PMNs and other myeloid cells in vitro, a function considered widely as the primary means by which PVL might contribute to disease. However, at sublytic concentrations, PVL can function as a PMN agonist. To better understand this phenomenon, we investigated the ability of PVL to alter human PMN function. PMNs exposed to PVL had enhanced capacity to produce O(2)(-) in response to fMLF, but unlike priming by LPS, this response did not require TLR signal transduction. On the other hand, there was subcellular redistribution of NADPH oxidase components in PMNs following exposure of these cells to PVL--a finding consistent with priming. Importantly, PMNs primed with PVL had an enhanced ability to bind/ingest and kill Staphylococcus aureus. Priming of PMNs with other agonists, such as IL-8 or GM-CSF, altered the ability of PVL to cause formation of pores in the plasma membranes of these cells. Microarray analysis revealed significant changes in the human PMN transcriptome following exposure to PVL, including up-regulation of molecules that regulate the inflammatory response. Consistent with the microarray data, mediators of the inflammatory response were released from PMNs after stimulation with PVL. We conclude that exposure of human PMNs to sublytic concentrations of PVL elicits a proinflammatory response that is regulated in part at the level of gene expression. We propose that PVL-mediated priming of PMNs enhances the host innate immune response.


Subject(s)
Exotoxins/physiology , Leukocidins/physiology , Methicillin-Resistant Staphylococcus aureus/immunology , Neutrophils/immunology , Neutrophils/microbiology , Staphylococcal Infections/immunology , Bacterial Toxins/metabolism , Cells, Cultured , Exotoxins/metabolism , Humans , Leukocidins/metabolism , Methicillin-Resistant Staphylococcus aureus/growth & development , Methicillin-Resistant Staphylococcus aureus/metabolism , Microbial Sensitivity Tests , Neutrophils/drug effects , Staphylococcal Infections/metabolism , Staphylococcal Infections/microbiology
6.
Infect Immun ; 80(3): 975-81, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22184421

ABSTRACT

Acetic acid bacteria were previously considered nonpathogenic in humans. However, over the past decade, five genera of Acetobacteraceae have been isolated from patients with inborn or iatrogenic immunodeficiencies. Here, we describe the first studies of the interactions of the human innate immune system with a member of this bacterial family, Granulibacter bethesdensis, an emerging pathogen in patients with chronic granulomatous disease (CGD). Efficient phagocytosis of G. bethesdensis by normal and CGD polymorphonuclear leukocytes (CGD PMN) required heat-labile serum components (e.g., C3), and binding of C3 and C9 to G. bethesdensis was detected by immunoblotting. However, this organism survived in human serum concentrations of ≥90%, indicating a high degree of serum resistance. Consistent with the clinical host tropism of G. bethesdensis, CGD PMN were unable to kill this organism, while normal PMN, in the presence of serum, reduced the number of CFU by about 50% after a 24-h coculture. This finding, together with the observations that G. bethesdensis was sensitive to H(2)O(2) but resistant to LL-37, a human cationic antimicrobial peptide, suggests an inherent resistance to O(2)-independent killing. Interestingly, 10 to 100 times greater numbers of G. bethesdensis were required to achieve the same level of reactive oxygen species (ROS) production induced by Escherichia coli in normal PMN. In addition to the relative inability of the organism to elicit production of PMN ROS, G. bethesdensis inhibited both constitutive and FAS-induced PMN apoptosis. These properties of reduced PMN activation and resistance to nonoxidative killing mechanisms likely play an important role in G. bethesdensis pathogenesis.


Subject(s)
Acetobacteraceae/immunology , Acetobacteraceae/pathogenicity , Gram-Negative Bacterial Infections/immunology , Gram-Negative Bacterial Infections/microbiology , Granulomatous Disease, Chronic/immunology , Granulomatous Disease, Chronic/microbiology , Immunity, Innate , Blood Bactericidal Activity , Colony Count, Microbial , Complement System Proteins/immunology , Escherichia coli/immunology , Humans , Microbial Viability , Neutrophils/immunology , Phagocytosis , Reactive Oxygen Species/metabolism
7.
Proc Natl Acad Sci U S A ; 108(44): 18091-6, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-22025717

ABSTRACT

Staphylococcus aureus is a bacterial pathogen known to cause infections in epidemic waves. One such epidemic was caused by a clone known as phage-type 80/81, a penicillin-resistant strain that rose to world prominence in the late 1950s. The molecular underpinnings of the phage-type 80/81 outbreak have remained unknown for decades, nor is it understood why related S. aureus clones became epidemic in hospitals in the early 1990s. To better understand the molecular basis of these epidemics, we sequenced the genomes of eight S. aureus clinical isolates representative of the phage-type 80/81 clone, the Southwest Pacific clone [a community-associated methicillin-resistant S. aureus (MRSA) clone], and contemporary S. aureus clones, all of which are genetically related and belong to the same clonal complex (CC30). Genome sequence analysis revealed that there was coincident divergence of these clones from a recent common ancestor, a finding that resolves controversy about the evolutionary history of the lineage. Notably, we identified nonsynonymous SNPs in genes encoding accessory gene regulator C (agrC) and α-hemolysin (hla)--molecules important for S. aureus virulence--that were present in virtually all contemporary CC30 hospital isolates tested. Compared with the phage-type 80/81 and Southwest Pacific clones, contemporary CC30 hospital isolates had reduced virulence in mouse infection models, the result of SNPs in agrC and hla. We conclude that agr and hla (along with penicillin resistance) were essential for world dominance of phage-type 80/81 S. aureus, whereas key SNPs in contemporary CC30 clones restrict these pathogens to hospital settings in which the host is typically compromised.


Subject(s)
Bacteriophages/classification , Staphylococcal Infections/epidemiology , Staphylococcus aureus/virology , Bacteriophages/genetics , Disease Outbreaks , Genome, Bacterial , Genome, Viral , Humans , Mutation , Phylogeny , Polymorphism, Single Nucleotide , Staphylococcus aureus/genetics , Staphylococcus aureus/pathogenicity , Virulence
8.
J Infect Dis ; 204(6): 937-41, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21849291

ABSTRACT

Community-associated methicillin-resistant Staphylococcus aureus (CA-MRSA) infections are frequently associated with strains harboring genes encoding Panton-Valentine leukocidin (PVL). The role of PVL in the success of the epidemic CA-MRSA strain USA300 remains unknown. Here we developed a skin and soft tissue infection model in rabbits to test the hypothesis that PVL contributes to USA300 pathogenesis and compare it with well-established virulence determinants: alpha-hemolysin (Hla), phenol-soluble modulin-alpha peptides (PSMα), and accessory gene regulator (Agr). The data indicate that Hla, PSMα, and Agr contribute to the pathogenesis of USA300 skin infections in rabbits, whereas a role for PVL could not be detected.


Subject(s)
Methicillin-Resistant Staphylococcus aureus/pathogenicity , Soft Tissue Infections/microbiology , Soft Tissue Infections/pathology , Staphylococcal Skin Infections/microbiology , Staphylococcal Skin Infections/pathology , Virulence Factors/metabolism , Abscess/microbiology , Abscess/pathology , Animals , Bacterial Load , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Toxins/genetics , Bacterial Toxins/metabolism , Disease Models, Animal , Exotoxins/genetics , Exotoxins/metabolism , Female , Hemolysin Proteins/genetics , Hemolysin Proteins/metabolism , Histocytochemistry , Leukocidins/genetics , Leukocidins/metabolism , Microscopy , Rabbits , Skin/microbiology , Skin/pathology , Trans-Activators/genetics , Trans-Activators/metabolism , Virulence , Virulence Factors/genetics
9.
PLoS One ; 6(4): e18617, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21525981

ABSTRACT

Staphylococcus aureus is a leading cause of bloodstream infections worldwide. In the United States, many of these infections are caused by a strain known as USA300. Although progress has been made, our understanding of the S. aureus molecules that promote survival in human blood and ultimately facilitate metastases is incomplete. To that end, we analyzed the USA300 transcriptome during culture in human blood, human serum, and trypticase soy broth (TSB), a standard laboratory culture media. Notably, genes encoding several cytolytic toxins were up-regulated in human blood over time, and hlgA, hlgB, and hlgC (encoding gamma-hemolysin subunits HlgA, HlgB, and HlgC) were among the most highly up-regulated genes at all time points. Compared to culture supernatants from a wild-type USA300 strain (LAC), those derived from an isogenic hlgABC-deletion strain (LACΔhlgABC) had significantly reduced capacity to form pores in human neutrophils and ultimately cause neutrophil lysis. Moreover, LACΔhlgABC had modestly reduced ability to cause mortality in a mouse bacteremia model. On the other hand, wild-type and LACΔhlgABC strains caused virtually identical abscesses in a mouse skin infection model, and bacterial survival and neutrophil lysis after phagocytosis in vitro was similar between these strains. Comparison of the cytolytic capacity of culture supernatants from wild-type and isogenic deletion strains lacking hlgABC, lukS/F-PV (encoding PVL), and/or lukDE revealed functional redundancy among two-component leukotoxins in vitro. These findings, along with a requirement of specific growth conditions for leukotoxin expression, may explain the apparent limited contribution of any single two-component leukotoxin to USA300 immune evasion and virulence.


Subject(s)
Gene Expression Regulation, Bacterial , Serum/microbiology , Staphylococcus aureus/genetics , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Toxins/genetics , Bacterial Toxins/metabolism , Cell Membrane Permeability , Disease Models, Animal , Exotoxins/genetics , Exotoxins/metabolism , Gene Deletion , Gene Expression Profiling , Hemolysin Proteins/genetics , Hemolysin Proteins/metabolism , Humans , Mice , Microbial Viability , Neutrophils/cytology , Neutrophils/microbiology , Porosity , RNA, Messenger/genetics , RNA, Messenger/metabolism , Staphylococcal Infections/genetics , Staphylococcal Infections/microbiology , Staphylococcus aureus/pathogenicity , Virulence/genetics
10.
PLoS Pathog ; 6(10): e1001133, 2010 Oct 07.
Article in English | MEDLINE | ID: mdl-20949069

ABSTRACT

Staphylococcus epidermidis is a leading nosocomial pathogen. In contrast to its more aggressive relative S. aureus, it causes chronic rather than acute infections. In highly virulent S. aureus, phenol-soluble modulins (PSMs) contribute significantly to immune evasion and aggressive virulence by their strong ability to lyse human neutrophils. Members of the PSM family are also produced by S. epidermidis, but their role in immune evasion is not known. Notably, strong cytolytic capacity of S. epidermidis PSMs would be at odds with the notion that S. epidermidis is a less aggressive pathogen than S. aureus, prompting us to examine the biological activities of S. epidermidis PSMs. Surprisingly, we found that S. epidermidis has the capacity to produce PSMδ, a potent leukocyte toxin, representing the first potent cytolysin to be identified in that pathogen. However, production of strongly cytolytic PSMs was low in S. epidermidis, explaining its low cytolytic potency. Interestingly, the different approaches of S. epidermidis and S. aureus to causing human disease are thus reflected by the adaptation of biological activities within one family of virulence determinants, the PSMs. Nevertheless, S. epidermidis has the capacity to evade neutrophil killing, a phenomenon we found is partly mediated by resistance mechanisms to antimicrobial peptides (AMPs), including the protease SepA, which degrades AMPs, and the AMP sensor/resistance regulator, Aps (GraRS). These findings establish a significant function of SepA and Aps in S. epidermidis immune evasion and explain in part why S. epidermidis may evade elimination by innate host defense despite the lack of cytolytic toxin expression. Our study shows that the strategy of S. epidermidis to evade elimination by human neutrophils is characterized by a passive defense approach and provides molecular evidence to support the notion that S. epidermidis is a less aggressive pathogen than S. aureus.


Subject(s)
Immune Evasion/physiology , Neutrophils/immunology , Staphylococcal Infections/immunology , Staphylococcus epidermidis/immunology , Staphylococcus epidermidis/pathogenicity , Amino Acid Sequence , Bacterial Toxins/chemistry , Bacterial Toxins/genetics , Bacterial Toxins/metabolism , Bacterial Toxins/pharmacology , Hemolysis/drug effects , Hemolysis/genetics , Humans , Immune Evasion/genetics , Immunity, Cellular/physiology , Molecular Sequence Data , Neutrophils/physiology , Phylogeny , Sequence Homology, Amino Acid , Staphylococcal Infections/complications , Staphylococcal Infections/microbiology , Staphylococcal Infections/pathology , Staphylococcus aureus/genetics , Staphylococcus aureus/immunology , Staphylococcus aureus/metabolism , Staphylococcus aureus/pathogenicity , Staphylococcus epidermidis/genetics , Staphylococcus epidermidis/metabolism
11.
J Clin Microbiol ; 48(12): 4504-11, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20943864

ABSTRACT

A community-associated methicillin-resistant Staphylococcus aureus (CA-MRSA) strain known as pulsed-field type USA300 (USA300) is epidemic in the United States. Previous comparative whole-genome sequencing studies demonstrated that there has been recent clonal emergence of a subset of USA300 isolates, which comprise the epidemic clone. Although the core genomes of these isolates are closely related, the level of diversity among USA300 plasmids was not resolved. Inasmuch as these plasmids might contribute to significant gene diversity among otherwise closely related USA300 isolates, we performed de novo sequencing of endogenous plasmids from 10 previously characterized USA300 clinical isolates obtained from different geographic locations in the United States. All isolates tested contained small (2- to 3-kb) and/or large (27- to 30-kb) plasmids. The large plasmids encoded heavy metal and/or antimicrobial resistance elements, including those that confer resistance to cadmium, bacitracin, macrolides, penicillin, kanamycin, and streptothricin, although all isolates were sensitive to minocycline, doxycycline, trimethoprim-sulfamethoxazole, vancomycin, teicoplanin, and linezolid. One of the USA300 isolates contained an archaic plasmid that encoded staphylococcal enterotoxins R, J, and P. Notably, the large plasmids (27 to 28 kb) from 8 USA300 isolates--those that comprise the epidemic USA300 clone--were virtually identical (99% identity) and similar to a large plasmid from strain USA300_TCH1516 (a previously sequenced USA300 strain from Houston, TX). These plasmids are largely divergent from the 37-kb plasmid of FPR3757, the first sequenced USA300 strain. The high level of plasmid sequence identity among the majority of closely related USA300 isolates is consistent with the recent clonal emergence hypothesis for USA300.


Subject(s)
DNA, Bacterial/genetics , Genetic Variation , Methicillin-Resistant Staphylococcus aureus/classification , Methicillin-Resistant Staphylococcus aureus/genetics , Plasmids , Staphylococcal Infections/microbiology , Anti-Bacterial Agents/pharmacology , Cluster Analysis , DNA, Bacterial/chemistry , Drug Resistance, Multiple, Bacterial , Humans , Metals, Heavy/toxicity , Molecular Epidemiology , Molecular Sequence Data , Phylogeny , Sequence Analysis, DNA , United States
12.
J Infect Dis ; 202(7): 1050-8, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20726702

ABSTRACT

Community-associated methicillin-resistant Staphylococcus aureus (CA-MRSA) infections are predominantly those affecting skin and soft tissues. Although progress has been made, our knowledge of the molecules that contribute to the pathogenesis of CA-MRSA skin infections is incomplete. We tested the hypothesis that alpha-hemolysin (Hla) contributes to the severity of USA300 skin infections in mice and determined whether vaccination against Hla reduces disease severity. Isogenic hla-negative (Deltahla) strains caused skin lesions in a mouse infection model that were significantly smaller than those caused by wild-type USA300 and Newman strains. Moreover, infection due to wild-type strains produced dermonecrotic skin lesions, whereas there was little or no dermonecrosis in mice infected with Deltahla strains. Passive immunization with Hla-specific antisera or active immunization with a nontoxigenic form of Hla significantly reduced the size of skin lesions caused by USA300 and prevented dermonecrosis. We conclude that Hla is a potential target for therapeutics or vaccines designed to moderate severe S. aureus skin infections.


Subject(s)
Bacterial Toxins/antagonists & inhibitors , Hemolysin Proteins/antagonists & inhibitors , Immunization, Passive/methods , Immunization/methods , Staphylococcal Skin Infections/immunology , Staphylococcus aureus/immunology , Animals , Disease Models, Animal , Female , Gene Deletion , Hemolysin Proteins/deficiency , Humans , Mice , Mice, Inbred BALB C , Skin/pathology , Staphylococcal Skin Infections/microbiology , Staphylococcus aureus/pathogenicity
13.
J Innate Immun ; 2(6): 560-75, 2010.
Article in English | MEDLINE | ID: mdl-20587998

ABSTRACT

Mechanisms underlying the enhanced virulence phenotype of community-associated methicillin-resistant Staphylococcus aureus (CA-MRSA) are incompletely defined, but presumably include evasion of killing by human polymorphonuclear leukocytes (PMNs or neutrophils). To better understand this phenomenon, we investigated the basis of rapid PMN lysis after phagocytosis of USA300, a prominent CA-MRSA strain. Survival of USA300 clinical isolates after phagocytosis ultimately resulted in neutrophil lysis. PMNs containing ingested USA300 underwent morphological changes consistent with apoptosis, but lysed rapidly thereafter (within 6 h), whereas cells undergoing FAS-mediated apoptosis or phagocytosis-induced cell death remained intact. Phagosome membranes remained intact until the point of PMN destruction, suggesting lysis was not caused by escape of S. aureus from phagosomes or the cytolytic action of pore-forming toxins. Microarray analysis of the PMN transcriptome after phagocytosis of representative community-associated S. aureus and healthcare-associated MRSA strains revealed changes unique to community-associated S. aureus strains, such as upregulation of transcripts involved in regulation of calcium homeostasis. Collectively, the data suggest that neutrophil destruction after phagocytosis of USA300 is in part a form of programmed necrosis rather than direct lysis by S. aureus pore-forming toxins. We propose that the ability of CA-MRSA strains to induce programmed necrosis of neutrophils is a component of enhanced virulence.


Subject(s)
Community-Acquired Infections/immunology , Methicillin-Resistant Staphylococcus aureus/immunology , Neutrophils/metabolism , Phagosomes/ultrastructure , Staphylococcal Infections/immunology , Apoptosis/genetics , Gene Expression Regulation/immunology , Humans , Membrane Transport Proteins/biosynthesis , Membrane Transport Proteins/genetics , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Microarray Analysis , Microscopy, Electron, Transmission , Necrosis/genetics , Neutrophils/immunology , Neutrophils/microbiology , Neutrophils/pathology , Phagocytosis , Species Specificity , Staphylococcal Infections/microbiology , Virulence Factors
14.
Proc Natl Acad Sci U S A ; 107(12): 5587-92, 2010 Mar 23.
Article in English | MEDLINE | ID: mdl-20231457

ABSTRACT

Community-associated methicillin-resistant Staphylococcus aureus (CA-MRSA) is epidemic in the United States, even rivaling HIV/AIDS in its public health impact. The pandemic clone USA300, like other CA-MRSA strains, expresses Panton-Valentine leukocidin (PVL), a pore-forming toxin that targets polymorphonuclear leukocytes (PMNs). PVL is thought to play a key role in the pathogenesis of necrotizing pneumonia, but data from rodent infection models are inconclusive. Rodent PMNs are less susceptible than human PMNs to PVL-induced cytolysis, whereas rabbit PMNs, like those of humans, are highly susceptible to PVL-induced cytolysis. This difference in target cell susceptibility could affect results of experimental models. Therefore, we developed a rabbit model of necrotizing pneumonia to compare the virulence of a USA300 wild-type strain with that of isogenic PVL-deletion mutant and -complemented strains. PVL enhanced the capacity of USA300 to cause severe lung necrosis, pulmonary edema, alveolar hemorrhage, hemoptysis, and death, hallmark clinical features of fatal human necrotizing pneumonia. Purified PVL instilled directly into the lung caused lung inflammation and injury by recruiting and lysing PMNs, which damage the lung by releasing cytotoxic granule contents. These findings provide insights into the mechanism of PVL-induced lung injury and inflammation and demonstrate the utility of the rabbit for studying PVL-mediated pathogenesis.


Subject(s)
Acute Lung Injury/etiology , Bacterial Toxins/toxicity , Exotoxins/toxicity , Leukocidins/toxicity , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Neutrophils/drug effects , Neutrophils/physiology , Pneumonia, Staphylococcal/etiology , Acute Lung Injury/microbiology , Acute Lung Injury/pathology , Animals , Bacterial Toxins/genetics , Disease Models, Animal , Exotoxins/genetics , Gene Deletion , Genes, Bacterial , Genetic Complementation Test , Humans , In Vitro Techniques , Leukocidins/genetics , Methicillin-Resistant Staphylococcus aureus/genetics , Neutrophils/pathology , Pneumonia, Staphylococcal/microbiology , Pneumonia, Staphylococcal/pathology , Rabbits , Virulence/genetics
15.
Proc Natl Acad Sci U S A ; 107(2): 888-93, 2010 Jan 12.
Article in English | MEDLINE | ID: mdl-20080771

ABSTRACT

Single-nucleotide changes are the most common cause of natural genetic variation among members of the same species, but there is remarkably little information bearing on how they alter bacterial virulence. We recently discovered a single-nucleotide mutation in the group A Streptococcus genome that is epidemiologically associated with decreased human necrotizing fasciitis ("flesh-eating disease"). Working from this clinical observation, we find that wild-type mtsR function is required for group A Streptococcus to cause necrotizing fasciitis in mice and nonhuman primates. Expression microarray analysis revealed that mtsR inactivation results in overexpression of PrsA, a chaperonin involved in posttranslational maturation of SpeB, an extracellular cysteine protease. Isogenic mutant strains that overexpress prsA or lack speB had decreased secreted protease activity in vivo and recapitulated the necrotizing fasciitis-negative phenotype of the DeltamtsR mutant strain in mice and monkeys. mtsR inactivation results in increased PrsA expression, which in turn causes decreased SpeB secreted protease activity and reduced necrotizing fasciitis capacity. Thus, a naturally occurring single-nucleotide mutation dramatically alters virulence by dysregulating a multiple gene virulence axis. Our discovery has broad implications for the confluence of population genomics and molecular pathogenesis research.


Subject(s)
Fasciitis, Necrotizing/genetics , Polymorphism, Single Nucleotide , Virulence/genetics , Animals , Fasciitis, Necrotizing/immunology , Fasciitis, Necrotizing/prevention & control , Genetic Variation , Humans , Macaca fascicularis/microbiology , Male , Mice , Neutrophils/physiology , Serotyping , Shock, Septic/microbiology , Streptococcus pyogenes/genetics , Up-Regulation
16.
Blood ; 115(8): 1519-29, 2010 Feb 25.
Article in English | MEDLINE | ID: mdl-20040766

ABSTRACT

We identified 18 patients with the distinct clinical phenotype of susceptibility to disseminated nontuberculous mycobacterial infections, viral infections, especially with human papillomaviruses, and fungal infections, primarily histoplasmosis, and molds. This syndrome typically had its onset in adulthood (age range, 7-60 years; mean, 31.1 years; median, 32 years) and was characterized by profound circulating monocytopenia (mean, 13.3 cells/microL; median, 14.5 cells/microL), B lymphocytopenia (mean, 9.4 cells/microL; median, 4 cells/microL), and NK lymphocytopenia (mean, 16 cells/microL; median, 5.5 cells/microL). T lymphocytes were variably affected. Despite these peripheral cytopenias, all patients had macrophages and plasma cells at sites of inflammation and normal immunoglobulin levels. Ten of these patients developed 1 or more of the following malignancies: 9 myelodysplasia/leukemia, 1 vulvar carcinoma and metastatic melanoma, 1 cervical carcinoma, 1 Bowen disease of the vulva, and 1 multiple Epstein-Barr virus(+) leiomyosarcoma. Five patients developed pulmonary alveolar proteinosis without mutations in the granulocyte-macrophage colony-stimulating factor receptor or anti-granulocyte-macrophage colony-stimulating factor autoantibodies. Among these 18 patients, 5 families had 2 generations affected, suggesting autosomal dominant transmission as well as sporadic cases. This novel clinical syndrome links susceptibility to mycobacterial, viral, and fungal infections with malignancy and can be transmitted in an autosomal dominant pattern.


Subject(s)
Genetic Diseases, Inborn/genetics , Genetic Predisposition to Disease/genetics , Leukopenia/genetics , Mycobacterium Infections/genetics , Mycoses/genetics , Myelodysplastic Syndromes/genetics , Papillomavirus Infections/genetics , Pedigree , Adolescent , Adult , Child , Female , Fungi , Genetic Diseases, Inborn/blood , Genetic Diseases, Inborn/complications , Humans , Leukocyte Count , Leukopenia/blood , Leukopenia/complications , Male , Middle Aged , Mycobacterium , Mycobacterium Infections/blood , Mycobacterium Infections/etiology , Mycoses/blood , Mycoses/etiology , Myelodysplastic Syndromes/blood , Myelodysplastic Syndromes/etiology , Neoplasms/blood , Neoplasms/etiology , Neoplasms/genetics , Papillomaviridae , Papillomavirus Infections/blood , Papillomavirus Infections/etiology
17.
J Infect Dis ; 200(5): 724-34, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19604047

ABSTRACT

sThe role of Panton-Valentine leukocidin (PVL) in Staphylococcus aureus pathogenesis is controversial. Here, we show that an unintended point mutation in the agr P2 promoter of S. aureus caused the phenotypes in gene regulation and murine pneumonia attributed to PVL by earlier investigators. In agreement with other studies that failed to detect similar effects of PVL using community-associated methicillin-resistant S. aureus strains, we found no significant effect of PVL on gene expression or pathogenesis after we repaired the mutation. These findings provide further evidence that PVL does not have a major impact on S. aureus pathogenesis. Moreover, our results demonstrate that a single nucleotide polymorphism in an intergenic region can dramatically affect bacterial physiology and virulence. Finally, our work emphasizes the need to frequently evaluate the integrity of the S. aureus agr locus.


Subject(s)
Bacterial Proteins/biosynthesis , Bacterial Toxins/biosynthesis , Exotoxins/biosynthesis , Leukocidins/biosynthesis , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Pneumonia, Staphylococcal/microbiology , Point Mutation , Promoter Regions, Genetic , Trans-Activators/biosynthesis , Virulence Factors/biosynthesis , Animals , Base Sequence , Body Weight , Community-Acquired Infections/microbiology , Gene Expression Regulation, Bacterial , Humans , Methicillin-Resistant Staphylococcus aureus/physiology , Mice , Molecular Sequence Data , Virulence
18.
PLoS One ; 4(4): e5210, 2009.
Article in English | MEDLINE | ID: mdl-19381294

ABSTRACT

It is becoming increasingly apparent that Staphylococcus aureus are able to survive engulfment by macrophages, and that the intracellular environment of these host cells, which is essential to innate host defenses against invading microorganisms, may in fact provide a refuge for staphylococcal survival and dissemination. Based on this, we postulated that S. aureus might induce cytoprotective mechanisms by changing gene expression profiles inside macrophages similar to obligate intracellular pathogens, such as Mycobacterium tuberculosis. To validate our hypothesis we first ascertained whether S. aureus infection could affect programmed cell death in human (hMDMs) and mouse (RAW 264.7) macrophages and, specifically, protect these cells against apoptosis. Our findings indicate that S. aureus-infected macrophages are more resistant to staurosporine-induced cell death than control cells, an effect partly mediated via the inhibition of cytochrome c release from mitochondria. Furthermore, transcriptome analysis of human monocyte-derived macrophages during S. aureus infection revealed a significant increase in the expression of antiapoptotic genes. This was confirmed by quantitative RT-PCR analysis of selected genes involved in mitochondria-dependent cell death, clearly showing overexpression of BCL2 and MCL1. Cumulatively, the results of our experiments argue that S. aureus is able to induce a cytoprotective effect in macrophages derived from different mammal species, which can prevent host cell elimination, and thus allow intracellular bacterial survival. Ultimately, it is our contention that this process may contribute to the systemic dissemination of S. aureus infection.


Subject(s)
Apoptosis/genetics , Macrophages/immunology , Phagocytosis , Staphylococcus aureus/immunology , Up-Regulation , Animals , Base Sequence , Blotting, Western , Cells, Cultured , DNA Primers , Flow Cytometry , Gene Expression , Humans , Mice , Microscopy, Fluorescence , Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
19.
J Infect Dis ; 199(11): 1698-706, 2009 Jun 01.
Article in English | MEDLINE | ID: mdl-19374556

ABSTRACT

Methicillin-resistant Staphylococcus aureus is problematic both in hospitals and in the community. Currently, we have limited understanding of mechanisms of innate immune evasion used by S. aureus. To that end, we created an isogenic deletion mutant in strain MW2 (USA400) of the saeR/S 2-component gene regulatory system and studied its role in mouse models of pathogenesis and during human neutrophil interaction. In this study, we demonstrate that saeR/S plays a distinct role in S. aureus pathogenesis and is vital for virulence of MW2 in a mouse model of sepsis. Moreover, deletion of saeR/S significantly impaired survival of MW2 in human blood and after neutrophil phagocytosis. Microarray analysis revealed that SaeR/S of MW2 influences expression of a wide variety of genes with diverse biological functions. These data provide new insight into how virulence is regulated in S. aureus and associates a specific staphylococcal gene-regulatory system with invasive staphylococcal disease.


Subject(s)
Bacterial Proteins/genetics , Immunity, Innate/genetics , Protein Kinases/genetics , Staphylococcal Infections/immunology , Staphylococcus aureus/genetics , Staphylococcus aureus/immunology , Animals , Disease Models, Animal , Mice , Mutagenesis , Neutrophils/microbiology , Neutrophils/physiology , Phagocytosis , Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Sepsis/microbiology , Sequence Deletion , Staphylococcus aureus/isolation & purification , Staphylococcus aureus/pathogenicity , Transcription Factors , Virulence
20.
J Immunol ; 181(10): 7138-46, 2008 Nov 15.
Article in English | MEDLINE | ID: mdl-18981135

ABSTRACT

Streptococcus pneumoniae naturally colonizes the nasopharynx as a commensal organism and sometimes causes infections in remote tissue sites. This bacterium is highly capable of resisting host innate immunity during nasopharyngeal colonization and disseminating infections. The ability to recruit complement factor H (FH) by S. pneumoniae has been implicated as a bacterial immune evasion mechanism against complement-mediated bacterial clearance because FH is a complement alternative pathway inhibitor. S. pneumoniae recruits FH through a previously defined FH binding domain of choline-binding protein A (CbpA), a major surface protein of S. pneumoniae. In this study, we show that CbpA binds to human FH, but not to the FH proteins of mouse and other animal species tested to date. Accordingly, deleting the FH binding domain of CbpA in strain D39 did not result in obvious change in the levels of pneumococcal bacteremia or virulence in a bacteremia mouse model. Furthermore, this species-specific pneumococcal interaction with FH was shown to occur in multiple pneumococcal isolates from the blood and cerebrospinal fluid. Finally, our phagocytosis experiments with human and mouse phagocytes and complement systems provide additional evidence to support our hypothesis that CbpA acts as a bacterial determinant for pneumococcal resistance to complement-mediated host defense in humans.


Subject(s)
Bacterial Proteins/metabolism , Neutrophils/immunology , Pneumococcal Infections/immunology , Animals , Bacterial Proteins/genetics , Blotting, Western , Cattle , Complement Factor H/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Horses , Humans , Mice , Mice, Inbred BALB C , Neutrophils/metabolism , Neutrophils/microbiology , Pneumococcal Infections/metabolism , Rabbits , Species Specificity , Streptococcus pneumoniae/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...