Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Front Cell Infect Microbiol ; 12: 898796, 2022.
Article in English | MEDLINE | ID: mdl-35909964

ABSTRACT

Calprotectin is a transition metal chelating protein of the innate immune response known to exert nutritional immunity upon microbial infection. It is abundantly released during inflammation and is therefore found at sites occupied by pathogens such as Pseudomonas aeruginosa and Staphylococcus aureus. The metal limitation induced by this protein has previously been shown to mediate P. aeruginosa and S. aureus co-culture. In addition to the transition metal sequestration role of calprotectin, it has also been shown to have metal-independent antimicrobial activity via direct cell contact. Therefore, we sought to assess the impact of this protein on the biofilm architecture of P. aeruginosa and S. aureus in monomicrobial and polymicrobial culture. The experiments described in this report reveal novel aspects of calprotectin's interaction with biofilm communities of P. aeruginosa and S. aureus discovered using scanning electron microscopy and confocal laser scanning microscopy. Our results indicate that calprotectin can interact with microbial cells by stimulating encapsulation in mesh-like structures. This physical interaction leads to compositional changes in the biofilm extracellular polymeric substance (EPS) in both P. aeruginosa and S. aureus.


Subject(s)
Biofilms , Immunity, Innate , Leukocyte L1 Antigen Complex , Pseudomonas aeruginosa , Staphylococcus aureus , Anti-Bacterial Agents/immunology , Anti-Bacterial Agents/pharmacology , Extracellular Polymeric Substance Matrix/genetics , Extracellular Polymeric Substance Matrix/immunology , Humans , Immunity, Innate/genetics , Immunity, Innate/immunology , Leukocyte L1 Antigen Complex/genetics , Leukocyte L1 Antigen Complex/immunology , Phagocytosis , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/immunology , Staphylococcus aureus/genetics , Staphylococcus aureus/immunology
2.
NPJ Biofilms Microbiomes ; 6(1): 55, 2020 11 27.
Article in English | MEDLINE | ID: mdl-33247129

ABSTRACT

Staphylococcus aureus is a prominent etiological agent of suppurative abscesses. In principle, abscess formation and purulent exudate are classical physiological features of healing and tissue repair. However, S. aureus deploys two coagulases that can usurp this classical host response and form distinct abscess lesions. Here, we establish that during coinfection with coagulase producers and non-producers, coagulases are shared public goods that contribute to staphylococcal persistence, abscess formation, and disease progression. Coagulase-negative mutants that do not produce the public goods themselves are able to exploit those cooperatively secreted by producers and thereby thrive during coinfection at the expense of others. This study shows the importance of social interactions among pathogens concerning clinical outcomes.


Subject(s)
Abscess/microbiology , Coagulase/genetics , Staphylococcal Infections/pathology , Staphylococcus aureus/pathogenicity , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Coagulase/metabolism , Disease Models, Animal , Gene Deletion , Humans , Mice , Microbial Interactions , Mutation , Staphylococcus aureus/genetics , Staphylococcus aureus/metabolism
3.
PLoS One ; 14(8): e0221565, 2019.
Article in English | MEDLINE | ID: mdl-31442275

ABSTRACT

Chronic wound infections are increasingly recognized to be dynamic and polymicrobial in nature, necessitating the development of wound models which reflect the complexities of infection in a non-healing wound. Wound slough isolated from human chronic wounds and transferred to mice was recently shown to create polymicrobial infection in mice, and there is potential this tool may be improved by cryogenic preservation. The purpose of this study was to investigate the application of cryogenic preservation to transferring polymicrobial communities, specifically by quantifying the effects of cryopreservation and wound microbiome transplantation. Slough from an established murine polymicrobial surgical excision model and five patients were subjected to three preservation strategies: refrigeration until infection, freezing in liquid nitrogen, or freezing in liquid nitrogen with glycerol solution prior to infection in individual mice. Four days following inoculation onto mice, wound microbiota were quantified using either culture isolation or by 16s rRNA gene community profiling and quantitative PCR. Cryogenic preservation did not significantly reduce bacterial viability. Reestablished microbial communities were significantly associated with patient of origin as well as host context (i.e., originally preserved from a patient versus mouse infection). Whereas preservation treatment did not significantly shape community composition, the transfers of microbiomes from human to mouse were characterized by reduced diversity and compositional changes. These findings indicated that changes should be expected to occur to community structure after colonization, and that compositional change is likely due to the rapid change in infection context as opposed to preservation strategy. Furthermore, species that were present in higher relative abundance in wound inoculate were more likely to colonize subsequent wounds, and wound inoculate with higher bacterial load established wound communities that were more compositionally similar. Results inform expectations for the complementation of chronic wound in vivo modeling with cryogenic preservation archives.


Subject(s)
Cryopreservation , Microbiota , Wounds and Injuries/microbiology , Animals , Bacterial Load , Cell Survival , Chronic Disease , Disease Models, Animal , Female , Freezing , Humans , Mice
4.
Proc Natl Acad Sci U S A ; 115(50): E11771-E11779, 2018 12 11.
Article in English | MEDLINE | ID: mdl-30463950

ABSTRACT

Coagulation is an innate defense mechanism intended to limit blood loss and trap invading pathogens during infection. However, Staphylococcus aureus has the ability to hijack the coagulation cascade and generate clots via secretion of coagulases. Although many S. aureus have this characteristic, some do not. The population dynamics regarding this defining trait have yet to be explored. We report here that coagulases are public goods that confer protection against antimicrobials and immune factors within a local population or community, thus promoting growth and virulence. By utilizing variants of a methicillin-resistant S. aureus we infer that the secretion of coagulases is a cooperative trait, which is subject to exploitation by invading mutants that do not produce the public goods themselves. However, overexploitation, "tragedy of the commons," does not occur at clinically relevant conditions. Our micrographs indicate this is due to spatial segregation and population viscosity. These findings emphasize the critical role of coagulases in a social evolution context and provide a possible explanation as to why the secretion of these public goods is maintained in mixed S. aureus communities.


Subject(s)
Coagulase/physiology , Methicillin-Resistant Staphylococcus aureus/enzymology , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Staphylococcal Infections/microbiology , Biofilms/growth & development , Blood Coagulation , Coagulase/genetics , Humans , Microbiota/genetics , Microbiota/physiology , Models, Biological , Mutation , Staphylococcal Infections/blood , Virulence
5.
Methods Mol Biol ; 1673: 213-225, 2018.
Article in English | MEDLINE | ID: mdl-29130176

ABSTRACT

The human mucosal environment in the gut is rich with interactions between microbiota and mammalian epithelia. Microbes such as the Gram-negative bacterium Pseudomonas aeruginosa may use quorum sensing to communicate with other microorganisms and mammalian cells to alter gene expression. Here, we present methodologies to evaluate the effects of P. aeruginosa N-(3-oxo-dodecanoyl)-L-homoserine lactone (3O-C12-HSL) on Caco-2 cell monolayers. First, we describe a method for assessing barrier function and permeability of epithelial cells when exposed to 3O-C12-HSL by measuring transepithelial electrical resistance (TER) and paracellular flow using fluorescently labeled dextran. Secondly, we detail methods to investigate the effect of 3O-C12-HSL on protein-protein interactions of epithelial junction proteins. Lastly, we will detail imaging techniques to visualize Caco-2 barrier disruption following exposure to 3O-C12-HSL through the use of confocal laser scanning microscopy (CLSM) and a super resolution technique, stimulated emission depletion (STED) microscopy, to achieve nanoscale visualization of Caco-2 monolayers.


Subject(s)
Epithelial Cells/drug effects , Homoserine/analogs & derivatives , Lactones/pharmacology , Mammals/metabolism , Animals , Caco-2 Cells , Cell Extracts , Electric Impedance , Electrophoresis, Polyacrylamide Gel , Epithelial Cells/metabolism , Fluorescence , Homoserine/pharmacology , Humans , Immunoblotting , Immunoprecipitation , Microscopy, Confocal , Tight Junctions/metabolism , ras GTPase-Activating Proteins/metabolism
6.
Anal Biochem ; 539: 144-148, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29107579

ABSTRACT

Monitoring patients with burn wounds for infection is standard practice because failure to rapidly and specifically identify a pathogen can result in poor clinical outcomes, including death. Therefore, a method that facilitates detection and identification of pathogens in situ within minutes of biopsy would be a significant benefit to clinicians. Mass spectrometry is rapidly becoming a standard tool in clinical settings, capable of identifying specific pathogens from complex samples. Imaging mass spectrometry (IMS) expands the information content by enabling spatial resolution of biomarkers in tissue samples as in histology, without the need for specific stains/antibodies. Herein, a murine model of thermal injury was used to study infection of burn tissue by Pseudomonas aeruginosa. This is the first use of IMS to detect P. aeruginosa infection in situ from thermally injured tissue. Multiple molecular features could be spatially resolved to infected or uninfected tissue. This demonstrates the potential use of IMS in a clinical setting to aid doctors in identifying both presence and species of pathogens in tissue.


Subject(s)
Biomarkers/analysis , Burns/microbiology , Pseudomonas aeruginosa/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Animals , Burns/complications , Burns/pathology , Carboxymethylcellulose Sodium/chemistry , Disease Models, Animal , Gelatin/chemistry , Mice , Optical Imaging , Pseudomonas Infections/complications , Pseudomonas Infections/microbiology
7.
mBio ; 8(2)2017 03 14.
Article in English | MEDLINE | ID: mdl-28292986

ABSTRACT

Environmental conditions affect bacterial behavior and can greatly influence the course of an infection. However, the environmental cues that elicit bacterial responses in specific infection sites are relatively unknown. Pseudomonas aeruginosa is ubiquitous in nature and typically innocuous. However, it is also one of the most prevalent causes of fatal sepsis in burn wound patients. The aim of this study was to determine the impact of environmental factors, specifically the availability of arginine, on the pathogenesis of P. aeruginosa in burn wound infections. Comparison of burned versus noninjured tissue revealed that l-arginine (l-Arg) was significantly depleted in burn wounds as a consequence of elevated arginase produced by myeloid-derived suppressor cells. We also observed that l-Arg was a potent chemoattractant for P. aeruginosa, and while low concentrations of l-Arg increased P. aeruginosa's swimming motility, high concentrations resulted in diminished swimming. Based on these observations, we tested whether the administration of exogenous l-Arg into the burn wound could attenuate the virulence of P. aeruginosa in thermally injured mice. Administration of l-Arg resulted in decreased P. aeruginosa spread and sepsis and increased animal survival. Taken together, these data demonstrate that the availability of environmental arginine greatly influences the virulence of P. aeruginosa in vivo and may represent a promising phenotype-modulating tool for future therapeutic avenues.IMPORTANCE Despite our growing understanding of the pathophysiology of burn wounds and the evolution of techniques and practices to manage infections, sepsis remains a significant medical concern for burn patients. P. aeruginosa continues to be a leader among all causes of bacteremic infections due to its tendency to cause complications in immunocompromised patients and its ubiquitous presence in the hospital setting. With the unforgiving emergence of multidrug-resistant strains, it is critical that alternative strategies to control or prevent septic infections in burn patients be developed in parallel with novel antimicrobial agents. In this study, we observed that administration of l-Arg significantly reduced bacterial spread and sepsis in burned mice infected with P. aeruginosa Given the safety of l-Arg in high doses and its potential wound-healing benefits, this conditionally essential amino acid may represent a useful tool to modulate bacterial behavior in vivo and prevent sepsis in burn patients.


Subject(s)
Arginine/metabolism , Burns/complications , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/growth & development , Pseudomonas aeruginosa/metabolism , Wound Infection/microbiology , Animals , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/metabolism , Arginine/administration & dosage , Disease Models, Animal , Mice , Pseudomonas Infections/drug therapy , Survival Analysis , Treatment Outcome , Virulence , Wound Infection/drug therapy
8.
Infect Immun ; 82(11): 4718-28, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25156721

ABSTRACT

In individuals with polymicrobial infections, microbes often display synergistic interactions that can enhance their colonization, virulence, or persistence. One of the most prevalent types of polymicrobial infection occurs in chronic wounds, where Pseudomonas aeruginosa and Staphylococcus aureus are the two most common causes. Although they are the most commonly associated microbial species in wound infections, very little is known about their interspecies relationship. Evidence suggests that P. aeruginosa-S. aureus coinfections are more virulent than monoculture infection with either species; however, difficulties in growing these two pathogens together in vitro have hampered attempts to uncover the mechanisms involved. Here we describe a simple and clinically relevant in vitro wound model that supported concomitant growth of P. aeruginosa and S. aureus. We observed that the ability of P. aeruginosa and S. aureus to survive antibiotic treatment increased when they were grown together in planktonic cocultures and that antibiotic tolerance was further enhanced when they were grown together in the wound model. We attributed this enhanced tolerance to both the "host-derived" and "bacterium-derived" matrix components. Taken together, our data indicate that P. aeruginosa and S. aureus may benefit each other by coinfecting wounds and that the host-derived matrix may serve as important a role as the bacterium-derived matrix in protecting bacteria from some antibiotics.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteriological Techniques/methods , Coculture Techniques/methods , Drug Resistance, Bacterial/physiology , Pseudomonas aeruginosa/drug effects , Staphylococcus aureus/drug effects , Pseudomonas aeruginosa/physiology , Staphylococcus aureus/physiology
9.
PLoS Genet ; 10(7): e1004518, 2014 Jul.
Article in English | MEDLINE | ID: mdl-25057820

ABSTRACT

Opportunistic infections caused by Pseudomonas aeruginosa can be acute or chronic. While acute infections often spread rapidly and can cause tissue damage and sepsis with high mortality rates, chronic infections can persist for weeks, months, or years in the face of intensive clinical intervention. Remarkably, this diverse infectious capability is not accompanied by extensive variation in genomic content, suggesting that the genetic capacity to be an acute or a chronic pathogen is present in most P. aeruginosa strains. To investigate the genetic requirements for acute and chronic pathogenesis in P. aeruginosa infections, we combined high-throughput sequencing-mediated transcriptome profiling (RNA-seq) and genome-wide insertion mutant fitness profiling (Tn-seq) to characterize gene expression and fitness determinants in murine models of burn and non-diabetic chronic wound infection. Generally we discovered that expression of a gene in vivo is not correlated with its importance for fitness, with the exception of metabolic genes. By combining metabolic models generated from in vivo gene expression data with mutant fitness profiles, we determined the nutritional requirements for colonization and persistence in these infections. Specifically, we found that long-chain fatty acids represent a major carbon source in both chronic and acute wounds, and P. aeruginosa must biosynthesize purines, several amino acids, and most cofactors during infection. In addition, we determined that P. aeruginosa requires chemotactic flagellar motility for fitness and virulence in acute burn wound infections, but not in non-diabetic chronic wound infections. Our results provide novel insight into the genetic requirements for acute and chronic P. aeruginosa wound infections and demonstrate the power of using both gene expression and fitness profiling for probing bacterial virulence.


Subject(s)
Brain Injuries/genetics , Gene Expression Profiling , Pseudomonas aeruginosa/genetics , Surgical Wound Infection/genetics , Animals , Brain Injuries/microbiology , Disease Models, Animal , Gene Expression Regulation, Bacterial , Genetic Fitness , High-Throughput Nucleotide Sequencing , Host-Pathogen Interactions/genetics , Humans , Mice , Opportunistic Infections/genetics , Opportunistic Infections/microbiology , Pseudomonas aeruginosa/pathogenicity , Surgical Wound Infection/microbiology , Virulence Factors/genetics
10.
Proc Natl Acad Sci U S A ; 111(21): 7819-24, 2014 May 27.
Article in English | MEDLINE | ID: mdl-24825893

ABSTRACT

The oral pathogen Aggregatibacter actinomycetemcomitans (Aa) resides in infection sites with many microbes, including commensal streptococci such as Streptococcus gordonii (Sg). During infection, Sg promotes the virulence of Aa by producing its preferred carbon source, l-lactate, a phenomenon referred to as cross-feeding. However, as with many streptococci, Sg also produces high levels of the antimicrobial hydrogen peroxide (H2O2), leading to the question of how Aa deals with this potent antimicrobial during coinfection. Here, we show that Aa possesses two complementary responses to H2O2: a detoxification or fight response mediated by catalase (KatA) and a dispersion or flight response mediated by Dispersin B (DspB), an enzyme that dissolves Aa biofilms. Using a murine abscess infection model, we show that both of these responses are required for Sg to promote Aa virulence. Although the role of KatA is to detoxify H2O2 during coinfection, 3D spatial analysis of mixed infections revealed that DspB is required for Aa to spatially organize itself at an optimal distance (>4 µm) from Sg, which we propose allows cross-feeding but reduces exposure to inhibitory levels of H2O2. In addition, these behaviors benefit not only Aa but also Sg, suggesting that fight and flight stimulate the fitness of the community. These results reveal that an antimicrobial produced by a human commensal bacterium enhances the virulence of a pathogenic bacterium by modulating its spatial location in the infection site.


Subject(s)
Aggregatibacter actinomycetemcomitans/pathogenicity , Bacterial Proteins/metabolism , Biofilms/growth & development , Catalase/metabolism , Coinfection/physiopathology , Glycoside Hydrolases/metabolism , Streptococcus gordonii/metabolism , Aggregatibacter actinomycetemcomitans/metabolism , Animals , Coinfection/microbiology , Hydrogen Peroxide/metabolism , Lactic Acid/metabolism , Mice , Microarray Analysis , Virulence
11.
Infect Immun ; 82(1): 92-100, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24126517

ABSTRACT

Diabetes affects 25.8 million people in the United States, or 8.3% of the population, and these numbers are even higher in developing countries. Diabetic patients are more susceptible to the development of chronic wounds with debilitating bacterial infections than nondiabetics. Previously, we compared the ability of the opportunistic pathogen Pseudomonas aeruginosa to cause biofilm-associated infections in chronic wounds of diabetic and nondiabetic mice (C. Watters, K. DeLeon, U. Trivedi, J. A. Griswold, M. Lyte, K. J. Hampel, M. J. Wargo, and K. P. Rumbaugh, Med. Microbiol. Immunol. 202:131-141, 2013). Unexpectedly, we observed that insulin-treated diabetic mice had significantly more biofilm in their wounds, which correlated with higher antibiotic tolerance. Here, we investigated whether insulin treatment modulates the diabetic immune system to favor P. aeruginosa biofilm formation. Utilizing a murine chronic wound model, we found that DNA protected P. aeruginosa in the wounds of insulin-treated diabetic mice from antibiotic treatment. We also observed increased numbers of neutrophils, reduced numbers of macrophages, and increased cell death in the wounds of diabetic mice on insulin therapy. Taken together, these data suggest that high levels of lysed neutrophils in the wounds of diabetic mice on insulin, combined with fewer macrophages to remove the cellular debris, contribute to increased DNA levels, which enhance P. aeruginosa biofilms.


Subject(s)
Biofilms/growth & development , Diabetes Mellitus, Experimental/immunology , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/drug effects , Wound Infection/microbiology , Animals , Anti-Bacterial Agents/pharmacology , Apoptosis/physiology , Cell Death/immunology , Chronic Disease , DNA, Bacterial/analysis , Diabetes Mellitus, Experimental/drug therapy , Disease Models, Animal , Drug Resistance, Bacterial/immunology , Female , Macrophages/cytology , Mice , Microbial Sensitivity Tests , Neutrophils/cytology , Pseudomonas Infections/complications , Pseudomonas Infections/pathology , Pseudomonas aeruginosa/physiology , Wound Healing/physiology , Wound Infection/complications , Wound Infection/pathology
SELECTION OF CITATIONS
SEARCH DETAIL