Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
J Orthop Res ; 35(9): 1845-1854, 2017 09.
Article in English | MEDLINE | ID: mdl-28387956

ABSTRACT

Extremity injuries comprise a significant portion of trauma, affecting quality of life, financial burden, and return to duty. Bacterial contamination is commonly associated with failure to heal, despite antibiotic treatment, suggesting that additional therapies must be developed to combat these complications. Treatment failure is likely due to the presence of resistant microbial communities known as biofilms. Biofilm bacteria are able to elicit a direct inhibition of healing through a multitude of known factors. However, they likely also inhibit healing through alteration of the inflammatory response. As inflammation is a critical step in fracture healing, how the presence of biofilm bacteria shifts this response to one that is suboptimal for healing is an important consideration that is currently understudied. The profile of inflammatory factors in response to biofilm bacteria is unique and distinct from those induced during normal healing or by planktonic bacteria alone. This review will examine the presence of inflammatory factors during normal healing and those induced by contaminating bacteria, and will discuss how these differences may ultimately lead to nonunion. Specifically, this review will focus on the Th1/Th2/Th17 type inflammatory responses and how shifts in the balance of these responses during infection can lead to both ineffective clearance and disruption of fracture healing. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 35:1845-1854, 2017.


Subject(s)
Biofilms , Fracture Healing/immunology , Staphylococcal Infections/immunology , Staphylococcus aureus/physiology , Wounds and Injuries/complications , Humans , Staphylococcal Infections/therapy
2.
PLoS One ; 8(4): e63040, 2013.
Article in English | MEDLINE | ID: mdl-23658662

ABSTRACT

Staphylococcus aureus is a major human pathogen and a leading cause of nosocomial and community-acquired infections. Development of a vaccine against this pathogen is an important goal. While S. aureus protective antigens have been identified in the literature, the majority have only been tested in a single animal model of disease. We wished to evaluate the ability of one S. aureus vaccine antigen to protect in multiple mouse models, thus assessing whether protection in one model translates to protection in other models encompassing the full breadth of infections the pathogen can cause. We chose to focus on genetically inactivated alpha toxin mutant HlaH35L. We evaluated the protection afforded by this antigen in three models of infection using the same vaccine dose, regimen, route of immunization, adjuvant, and challenge strain. When mice were immunized with HlaH35L and challenged via a skin and soft tissue infection model, HlaH35L immunization led to a less severe infection and decreased S. aureus levels at the challenge site when compared to controls. Challenge of HlaH35L-immunized mice using a systemic infection model resulted in a limited, but statistically significant decrease in bacterial colonization as compared to that observed with control mice. In contrast, in a prosthetic implant model of chronic biofilm infection, there was no significant difference in bacterial levels when compared to controls. These results demonstrate that vaccines may confer protection against one form of S. aureus disease without conferring protection against other disease presentations and thus underscore a significant challenge in S. aureus vaccine development.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Toxins/immunology , Soft Tissue Infections/prevention & control , Staphylococcal Infections/prevention & control , Staphylococcal Skin Infections/prevention & control , Staphylococcal Vaccines/immunology , Adjuvants, Immunologic , Animals , Antibodies, Bacterial/immunology , Antigens, Bacterial/administration & dosage , Antigens, Bacterial/genetics , Bacterial Toxins/administration & dosage , Bacterial Toxins/genetics , Biofilms/drug effects , Biofilms/growth & development , Colony Count, Microbial , Disease Models, Animal , Female , Immunization , Immunoglobulin G/immunology , Mice , Soft Tissue Infections/immunology , Soft Tissue Infections/microbiology , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Staphylococcal Skin Infections/immunology , Staphylococcal Skin Infections/microbiology , Staphylococcal Vaccines/administration & dosage , Staphylococcal Vaccines/genetics , Staphylococcus aureus/drug effects , Staphylococcus aureus/growth & development , Staphylococcus aureus/immunology , Vaccines, Attenuated
3.
PLoS One ; 8(3): e59232, 2013.
Article in English | MEDLINE | ID: mdl-23555002

ABSTRACT

In vivo bioluminescent imaging permits the visualization of bacteria in live animals, allowing researchers to monitor, both temporally and spatially, the progression of infection in each animal. We sought to engineer stably luminescent clinical strains of Staphylococcus aureus, with the goal of using such strains in mouse models. The gram-positive shuttle vector pMAD was used as the backbone for an integration plasmid. A chloramphenicol resistance gene, a modified lux operon from Photorhabdus luminescens, and approximately 650 bp of homology to the chromosome of the USA300 S. aureus strain NRS384 were added, generating plasmid pRP1195. Electroporation into strain RN4220 followed by temperature shift led to integration of pRP1195 into the chromosome. The integrated plasmid was transferred to clinical strains by phage transduction. Luminescent strains displayed no in vitro growth defects. Moreover, luminescence was stable in vitro after three rounds of subculture over 48 hours of growth in the absence of antibiotics. Mice were infected with a luminescent strain of NRS384 in skin and intravenous models. In a mouse skin model, luminescent bacteria were present in lesions that formed and cleared over the course of several days, and in an intravenous model, bacteria inoculated in the mouse tail vein were observed spreading to multiple tissues. No statistically significant difference in virulence was observed between NRS384 and the luminescent strain in either infection model. These preliminary data suggest that this luminescent USA300 strain is suitable for use in mouse models. Similar strains were engineered using other sequenced clinical strains. Because these strains are stably luminescent, they should prove useful in animal models of infection.


Subject(s)
Chromosomes, Bacterial/genetics , Founder Effect , Gene Transfer Techniques , Plasmids , Staphylococcal Infections/microbiology , Staphylococcus aureus/genetics , Animals , Bacteriophages/genetics , Electroporation , Injections, Intravenous , Luminescence , Luminescent Measurements , Mice , Operon , Photorhabdus/chemistry , Photorhabdus/genetics , Staphylococcal Infections/mortality , Staphylococcus aureus/isolation & purification , Staphylococcus aureus/ultrastructure , Survival Analysis
4.
Infect Immun ; 81(4): 1306-15, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23381997

ABSTRACT

Staphylococcus aureus is one of the most common etiological agents of community-acquired skin and soft tissue infection (SSTI). Although the majority of S. aureus community-acquired SSTIs are uncomplicated and self-clearing in nature, some percentage of these cases progress into life-threatening invasive infections. Current animal models of S. aureus SSTI suffer from two drawbacks: these models are a better representation of hospital-acquired SSTI than community-acquired SSTI, and they involve methods that are difficult to replicate. For these reasons, we sought to develop a murine model of community-acquired methicillin-resistant S. aureus SSTI (CA-MRSA SSTI) that can be consistently reproduced with a high degree of precision. We utilized this model to begin to characterize the host immune response to this type of infection. We infected mice via epicutaneous challenge of the skin on the outer ear pinna using Morrow-Brown allergy test needles coated in S. aureus USA300. When mice were challenged in this model, they developed small, purulent, self-clearing lesions with predictable areas of inflammation that mimicked a human infection. CFU in the ear pinna peaked at day 7 before dropping by day 14. The T(h)1 and T(h)17 cytokines gamma interferon (IFN-γ), interleukin-12 (IL-12) p70, tumor necrosis factor alpha (TNF-α), IL-17A, IL-6, and IL-21 were all significantly increased in the draining lymph node of infected mice, and there was neutrophil recruitment to the infection site. In vivo neutrophil depletion demonstrated that neutrophils play a protective role in preventing bacterial dissemination and fatal invasive infection.


Subject(s)
Community-Acquired Infections/microbiology , Community-Acquired Infections/pathology , Disease Models, Animal , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Staphylococcal Skin Infections/microbiology , Staphylococcal Skin Infections/pathology , Animals , Bacterial Load , Cytokines/analysis , Cytokines/immunology , Ear, External/microbiology , Ear, External/pathology , Female , Lymph Nodes/chemistry , Lymph Nodes/immunology , Mice , Mice, Inbred BALB C , Neutrophils/immunology , Skin/microbiology , Skin/pathology , Time Factors
5.
Infect Immun ; 79(12): 5010-8, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21947772

ABSTRACT

Staphylococcus aureus is a common cause of prosthetic implant infections, which can become chronic due to the ability of S. aureus to grow as a biofilm. Little is known about adaptive immune responses to these infections in vivo. We hypothesized that S. aureus elicits inflammatory Th1/Th17 responses, associated with biofilm formation, instead of protective Th2/Treg responses. We used an adapted mouse model of biofilm-mediated prosthetic implant infection to determine chronic infection rates, Treg cell frequencies, and local cytokine levels in Th1-biased C57BL/6 and Th2-biased BALB/c mice. All C57BL/6 mice developed chronic S. aureus implant infection at all time points tested. However, over 75% of BALB/c mice spontaneously cleared the infection without adjunctive therapy and demonstrated higher levels of Th2 cytokines and anti-inflammatory Treg cells. When chronic infection rates in mice deficient in the Th2 cytokine interleukin-4 (IL-4) via STAT6 mutation in a BALB/c background were assessed, the mice were unable to clear the S. aureus implant infection. Additionally, BALB/c mice depleted of Treg cells via an anti-CD25 monoclonal antibody (MAb) were also unable to clear the infection. In contrast, the C57BL/6 mice that were susceptible to infection were able to eliminate S. aureus biofilm populations on infected intramedullary pins once the Th1 and Th17 responses were diminished by MAb treatment with anti-IL-12 p40. Together, these results indicate that Th2/Treg responses are mechanisms of protection against chronic S. aureus implant infection, as opposed to Th1/Th17 responses, which may play a role in the development of chronic infection.


Subject(s)
Biofilms/growth & development , Inflammation/prevention & control , Methicillin-Resistant Staphylococcus aureus/physiology , Staphylococcal Infections/immunology , Animals , Antibodies, Monoclonal/therapeutic use , Chronic Disease , Immunity, Innate , Interleukin-12 Subunit p40/immunology , Interleukin-6/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Prostheses and Implants/microbiology , STAT6 Transcription Factor/genetics , STAT6 Transcription Factor/metabolism , Staphylococcal Infections/microbiology , T-Lymphocytes, Regulatory/immunology , Th2 Cells/physiology
6.
Infect Immun ; 79(4): 1789-96, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21282411

ABSTRACT

Staphylococcus aureus has reemerged as an important human pathogen in recent decades. Although many infections caused by this microbial species persist through a biofilm mode of growth, little is known about how the host's adaptive immune system responds to these biofilm infections. In this study, S. aureus cells adhered to pins in culture and were subsequently inserted into the tibiae of C57BL/6 mice, with an infecting dose of 2 × 105 CFU. This model was utilized to determine local cytokine levels, antibody (Ab) function, and T cell populations at multiple time points throughout infection. Like human hosts, S. aureus implant infection was chronic and remained localized in 100% of C57BL/6 mice at a consistent level of approximately 10(7) CFU/gram bone tissue after day 7. This infection persisted locally for >49 days and was recalcitrant to clearance by the host immune response and antimicrobial therapy. Local inflammatory cytokines of the Th1 (interleukin-2 [IL-2], IL-12 p70, tumor necrosis factor alpha [TNF-α], and IL-1ß) and Th17 (IL-6 and IL-17) responses were upregulated throughout the infection, except IL-12 p70, which dwindled late in the infection. In addition, Th1 Ab subtypes against a biofilm antigen (SA0486) were upregulated early in the infection, while Th2 Abs and anti-inflammatory regulatory T cells (Tregs) were not upregulated until later. These results indicate that early Th1 and Th17 inflammatory responses and downregulated Th2 and Treg responses occur during the development of a chronic biofilm implant infection. This unrestrained inflammatory response may cause tissue damage, thereby enabling S. aureus to attach and thrive in a biofilm mode of growth.


Subject(s)
Biofilms/growth & development , Bone Nails/microbiology , Prosthesis-Related Infections/immunology , Staphylococcal Infections/immunology , Staphylococcus aureus/physiology , Animals , Chronic Disease , Cytokines/immunology , Flow Cytometry , Immunoglobulin G/blood , Immunoglobulin G/immunology , Mice , Mice, Inbred C57BL , T-Lymphocytes/immunology
7.
J Orthop Trauma ; 24(9): 583-6, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20736799

ABSTRACT

Musculoskeletal infection is a clinical problem with significant direct healthcare costs. The prevalence of infection after closed, elective surgery is frequently estimated to be less than 2%, but in severe injuries, posttraumatic infection rates have been reported as 10% or greater. Although clinical infections are found outside the realm of medical devices, it is clear that the enormous increase of infections associated with the use of implants presents a major challenge worldwide. This review summarizes recent advances in the understanding, diagnosis, and treatment of musculoskeletal infections.


Subject(s)
Bacterial Infections/diagnosis , Musculoskeletal Diseases/diagnosis , Prostheses and Implants/microbiology , Prosthesis-Related Infections/diagnosis , Surgical Wound Infection/diagnosis , Animals , Bacterial Infections/microbiology , Biofilms , Disease Models, Animal , Drug Resistance, Microbial , Host-Pathogen Interactions , Humans , Musculoskeletal Diseases/microbiology , Prosthesis-Related Infections/microbiology , Surgical Wound Infection/microbiology
8.
FEMS Immunol Med Microbiol ; 59(3): 306-23, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20602638

ABSTRACT

Vaccine development against pathogenic bacteria is an imperative initiative as bacteria are gaining resistance to current antimicrobial therapies and few novel antibiotics are being developed. Candidate antigens for vaccine development can be identified by a multitude of high-throughput technologies that were accelerated by access to complete genomes. While considerable success has been achieved in vaccine development against bacterial pathogens, many species with multiple virulence factors and modes of infection have provided reasonable challenges in identifying protective antigens. In particular, vaccine candidates should be evaluated in the context of the complex disease properties, whether planktonic (e.g. sepsis and pneumonia) and/or biofilm associated (e.g. indwelling medical device infections). Because of the phenotypic differences between these modes of growth, those vaccine candidates chosen only for their efficacy in one disease state may fail against other infections. This review will summarize the history and types of bacterial vaccines and adjuvants as well as present an overview of modern antigen discovery and complications brought about by polymicrobial infections. Finally, we will also use one of the better studied microbial species that uses differential, multifactorial protein profiles to mediate an array of diseases, Staphylococcus aureus, to outline some of the more recently identified problematic issues in vaccine development in this biofilm-forming species.


Subject(s)
Biofilms/growth & development , Staphylococcal Infections/microbiology , Staphylococcal Infections/prevention & control , Staphylococcal Vaccines/immunology , Staphylococcus aureus/immunology , Staphylococcus aureus/physiology , Antigens, Bacterial/immunology , Humans , Staphylococcal Infections/immunology , Staphylococcus aureus/pathogenicity
9.
J Virol ; 76(10): 4810-21, 2002 May.
Article in English | MEDLINE | ID: mdl-11967298

ABSTRACT

There is currently intensive research on the design of novel human immunodeficiency virus type 1 (HIV-1) vaccine immunogens that can elicit potent neutralizing antibodies. A prerequisite for comparing and optimizing these strategies is the ability to precisely measure neutralizing antibody responses. To this end, we sought to develop an assay that directly quantifies single-round HIV-1 infection of peripheral blood mononuclear cells (PBMC). Initial experiments demonstrated that essentially all productively infected PBMC could be identified by flow cytometric detection of intracellular p24 antigen (p24-Ag). After infection of PBMC with HIV-1, p24(+) lymphocytes could be distinguished beginning 1 day postinfection, and the majority of CD8(-) T cells were p24-Ag positive by 3 to 4 days postinfection. To directly quantify first-round infection, we included a protease inhibitor in PBMC cultures. The resulting 2-day assay was highly sensitive and specific for the detection of HIV-1-infected PBMC. Serial dilutions of virus stocks demonstrated that the number of target cells infected was directly related to the amount of infectious virus input into the assay. In neutralization assays, the flow cytometric enumeration of first-round infection of PBMC provided quantitative data on the number of target cells infected and on the inactivation of infectious virus due to reaction with antibody. We also used this single-round assay to compare the percentage of cells expressing p24-Ag to the number of copies of HIV-1 gag per 100 PBMC. The precision and reproducibility of this assay will facilitate the measurement of HIV-1 neutralization, particularly incrementally improved neutralizing antibody responses generated by new candidate vaccines.


Subject(s)
HIV-1/immunology , Neutralization Tests/methods , T-Lymphocytes/virology , Cells, Cultured , Flow Cytometry , HIV Antibodies/immunology , HIV Core Protein p24/analysis , HIV-1/isolation & purification , Humans , Leukocytes, Mononuclear/virology , Predictive Value of Tests , Protease Inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...