Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
J Immunol ; 185(7): 4272-83, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20826748

ABSTRACT

Pseudomonas aeruginosa is a major cause of blindness and visual impairment in the United States and worldwide. Using a murine model of keratitis in which abraded corneas are infected with P. aeruginosa parent and ΔfliC (aflagellar) strains 19660 and PAO1, we found that F4/80(+) macrophages were the predominant cell type in the cornea expressing TLR2, TLR4, and TLR5. Depletion of macrophages and dendritic cells using transgenic Mafia mice, in which Fas ligand is selectively activated in these cells, resulted in diminished cytokine production and cellular infiltration to the corneal stroma and unimpaired bacterial growth. TLR4(-/-) mice showed a similar phenotype postinfection with ΔfliC strains, whereas TLR4/5(-/-) mice were susceptible to corneal infection with parent strains. Bone marrow-derived macrophages stimulated with ΔfliC bacteria induced Toll/IL-1R intracellular domain (TIR)-containing adaptor inducing IFN-ß (TRIF)-dependent phosphorylation of IFN regulatory factor 3 in addition to TIR-containing adaptor protein/MyD88-dependent phosphorylation of IκB and nuclear translocation of the p65 subunit of NFκB. Furthermore, TRIF(-/-) mice showed a similar phenotype as TLR4(-/-) mice in regulating only ΔfliC bacteria, whereas MyD88(-/-) mice were unable to clear parent or ΔfliC bacteria. Finally, IL-1R1(-/-) and IL-1α/ß(-/-) mice were highly susceptible to infection. Taken together, these findings indicate that P. aeruginosa activates TLR4/5 on resident corneal macrophages, which signal through TRIF and TIR-containing adaptor protein/MyD88 pathways, leading to NF-κB translocation to the nucleus, transcription of CXCL1 and other CXC chemokines, recruitment of neutrophils to the corneal stroma, and subsequent bacterial killing and tissue damage. IL-1α and IL-1ß are also produced, which activate an IL-1R1/MyD88-positive feedback loop in macrophages and IL-1R on other resident cells in the cornea.


Subject(s)
Keratitis/immunology , Macrophages/immunology , Myeloid Differentiation Factor 88/immunology , Pseudomonas Infections/immunology , Toll-Like Receptor 4/immunology , Toll-Like Receptor 5/immunology , Adaptor Proteins, Vesicular Transport/immunology , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Blotting, Western , Cell Separation , Cornea/immunology , Cornea/metabolism , Cornea/microbiology , Disease Models, Animal , Flow Cytometry , Immunohistochemistry , Keratitis/microbiology , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Confocal , Myeloid Differentiation Factor 88/metabolism , Polymerase Chain Reaction , Pseudomonas Infections/metabolism , Pseudomonas aeruginosa/immunology , Signal Transduction/immunology , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 5/metabolism
2.
Adv Ther ; 26(4): 447-54, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19381523

ABSTRACT

INTRODUCTION: Antibacterial activity of ophthalmic fourth-generation fluoroquinolones has traditionally been evaluated by comparing only their active ingredients, gatifloxacin and moxifloxacin. However, ophthalmic formulations of fourth-generation fluoroquinolones differ in terms of the inclusion of preservatives. While gatifloxacin ophthalmic solution 0.3% (Zymar; Allergan, Inc., Irvine, CA, USA) contains 0.005% benzalkonium chloride (BAK), moxifloxacin ophthalmic solution 0.5% (Vigamox; Alcon Laboratories, Inc., Fort Worth, TX, USA) is preservative-free. Recent studies have demonstrated that the presence of BAK dramatically affects the antibacterial activity of the ophthalmic formulation of gatifloxacin. This study was designed to compare the kill rates of ophthalmic solutions of fourth-generation fluoroquinolones against isolates of common ocular bacterial pathogens. METHODS: Approximately 5.6 log(10) colony-forming units (CFU)/mL of Haemophilus influenzae (n=1), Streptococcus pneumoniae (n=1), Staphylococcus aureus (n=2), methicillin-resistant Staphylococcus aureus (MRSA) (n=4), methicillinresistant Staphylococcus epidermidis (MRSE) (n=4), and fluoroquinolone-resistant S. epidermidis (n=1) were incubated with ophthalmic solutions of either gatifloxacin or moxifloxacin. Viable bacteria were quantified at specific time points up to 60 minutes. RESULTS: Gatifloxacin 0.3% completely eradicated H. influenzae and Strep. pneumoniae in 5 minutes, one of two S. aureus isolates in 15 minutes, and the other S. aureus isolate in 60 minutes. Gatifloxacin 0.3% completely killed all MRSA, MRSE, and fluoroquinolone-resistant S. epidermidis isolates in 15 minutes. Moxifloxacin 0.5% completely eradicated Strep. pneumoniae and one of four MRSA isolates in 60 minutes. All other isolates incubated with moxifloxacin 0.5% retained viable bacteria ranging from 1.8 to 4.4 log(10) CFU/mL. CONCLUSIONS: The ophthalmic solution of gatifloxacin 0.3% eradicated bacteria that frequently cause postoperative ocular infections substantially faster than did the ophthalmic solution of moxifloxacin 0.5%.


Subject(s)
Anti-Infective Agents/therapeutic use , Aza Compounds/therapeutic use , Eye Infections, Bacterial/microbiology , Fluoroquinolones/therapeutic use , Ophthalmic Solutions/therapeutic use , Quinolines/therapeutic use , Anti-Infective Agents/chemistry , Aza Compounds/chemistry , Benzalkonium Compounds/therapeutic use , Colony Count, Microbial , Drug Evaluation, Preclinical , Drug Resistance, Bacterial , Endophthalmitis/microbiology , Eye Infections, Bacterial/prevention & control , Fluoroquinolones/chemistry , Gatifloxacin , Haemophilus influenzae/drug effects , Humans , Keratitis/microbiology , Moxifloxacin , Ophthalmic Solutions/chemistry , Postoperative Complications/microbiology , Preservatives, Pharmaceutical/therapeutic use , Quinolines/chemistry , Staphylococcus aureus/drug effects , Staphylococcus epidermidis/drug effects , Streptococcus pneumoniae/drug effects , Time Factors
3.
Invest Ophthalmol Vis Sci ; 50(8): 3783-93, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19264886

ABSTRACT

PURPOSE: The purpose of this study was to determine to what extent blood-retinal barrier (BRB) permeability occurred during experimental Bacillus cereus endophthalmitis and whether tight junction alterations were involved in permeability. METHODS: Mice were intravitreally injected with 100 colony-forming units of B. cereus, and eyes were analyzed at specific times after infection for permeability to fibrin and albumin, quantitation of intraocular plasma constituent leakage, production of inflammatory cytokines, and alterations in tight junction protein localization and expression at the level of the retinal pigment epithelium. RESULTS: B. cereus induced the leakage of albumin and fibrin into the aqueous and vitreous humor by 8 hours after infection. BRB permeability occurred as early as 4 hours and increased 13.30-fold compared with uninfected controls by 8 hours. Production of proinflammatory cytokines IL-6, MIP-1alpha, IL-1beta, and KC increased over the course of infection. In the retina, ZO-1 disruption began by 4 hours and was followed by decreasing occludin and ZO-1 expression at 4 and 8 hours, respectively. Tubulin condensation and RPE65 degradation occurred by 12 hours. A quorum-sensing mutant B. cereus strain caused BRB permeability comparable to that of wild-type B. cereus. Wild-type and mutant B. cereus sterile supernatants induced blood-ocular barrier permeability similarly to that of wild-type infection. CONCLUSIONS: These results indicate that BRB permeability occurs during the early stages of experimental B. cereus endophthalmitis, beginning as early as 4 hours after infection. Disruption of tight junctions at the level of the retinal pigment epithelium may contribute to barrier breakdown. Quorum-sensing dependent factors may not significantly contribute to BRB permeability.


Subject(s)
Bacillus cereus/physiology , Blood-Retinal Barrier/microbiology , Capillary Permeability/physiology , Endophthalmitis/microbiology , Gram-Positive Bacterial Infections/microbiology , Animals , Aqueous Humor/metabolism , Blotting, Western , Cytokines/metabolism , Endophthalmitis/metabolism , Endophthalmitis/pathology , Fibrin/metabolism , Fluorescent Antibody Technique, Indirect , Gram-Positive Bacterial Infections/metabolism , Gram-Positive Bacterial Infections/pathology , Male , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Occludin , Phosphoproteins/metabolism , Serum Albumin/metabolism , Tight Junctions/metabolism , Vitreous Body/metabolism , Zonula Occludens-1 Protein
4.
Invest Ophthalmol Vis Sci ; 49(10): 4482-9, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18586878

ABSTRACT

PURPOSE: To determine the contribution of tumor necrosis factor-alpha (TNFalpha) in the pathogenesis of experimental Bacillus cereus endophthalmitis. METHODS: Experimental B. cereus endophthalmitis was induced in wild-type control (B6.129F1) and age-matched homozygous TNFalpha knockout mice (TNFalpha(-/-), B6.129S6-Tnf(tm1Gk1)/J). At various times after infection, eyes were analyzed by electroretinography and were harvested for quantitation of bacteria, myeloperoxidase, proinflammatory cytokines and chemokines, and histologic analysis. RESULTS: B. cereus replicated more rapidly in the eyes of TNFalpha(-/-) mice than in the eyes of B6.129F1 mice. Retinal function decreased more rapidly in TNFalpha(-/-) mice than in B6.129F1 mice. Retinal layers were not as structurally intact at 6 and 12 hours after infection in TNFalpha(-/-) eyes as in B6.129F1 eyes. Histologic analysis suggested less polymorphonuclear leukocyte (PMN) infiltration into the vitreous of TNFalpha(-/-) mice than of B6.129F1 mice. B6.129F1 eyes also had greater myeloperoxidase concentrations than did eyes of TNFalpha(-/-) mice. In general, concentrations of proinflammatory cytokines and chemokines (IL-1beta, KC, IL-6, and MIP-1alpha) were greater in eyes of TNFalpha(-/-) mice than of B6.129F1 mice. CONCLUSIONS: TNFalpha is important to intraocular pathogen containment by PMNs during experimental B. cereus endophthalmitis. In the absence of TNFalpha, fewer PMNs migrated into the eye, facilitating faster bacterial replication and retinal function loss. Although greater concentrations of proinflammatory cytokines were synthesized in the absence of TNFalpha, the resultant inflammation was diminished, and an equally devastating course of infection occurred.


Subject(s)
Bacillus cereus/physiology , Endophthalmitis/microbiology , Eye Infections, Bacterial/microbiology , Gram-Positive Bacterial Infections/microbiology , Tumor Necrosis Factor-alpha/physiology , Animals , Colony Count, Microbial , Cytokines/metabolism , Electroretinography , Endophthalmitis/immunology , Endophthalmitis/physiopathology , Eye Infections, Bacterial/immunology , Eye Infections, Bacterial/physiopathology , Female , Gram-Positive Bacterial Infections/immunology , Gram-Positive Bacterial Infections/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/immunology , Peroxidase/metabolism , Polymerase Chain Reaction , Retina/physiopathology , Vitreous Body/immunology , Vitreous Body/microbiology
5.
Prog Retin Eye Res ; 26(2): 189-203, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17236804

ABSTRACT

Endophthalmitis is an infection of the posterior segment of the eye that frequently results in loss of vision. This devastating result occurs despite prompt and often aggressive therapeutic and surgical intervention. Over the past decade, research has centered on determining the bacterial and host factors involved in this potentially blinding disease. The initial focus on the bacterial factors responsible for intraocular virulence has recently expanded into analysis the inflammatory response to infection, including the molecular and cellular interactions between the pathogen and host. This review discusses the epidemiology and therapeutic challenges posed by endophthalmitis, as well as recent findings from the analysis of interactions between the host and pathogen. Based on these findings, a model for the pathogenesis of endophthalmitis is presented. A more comprehensive understanding of the molecular and cellular interactions taking place between pathogen and host during endophthalmitis will expose possible therapeutic targets designed to arrest the infection and prevent vision loss.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Bacteria/pathogenicity , Bacterial Physiological Phenomena , Endophthalmitis/drug therapy , Endophthalmitis/microbiology , Eye Infections, Bacterial/drug therapy , Eye Infections, Bacterial/microbiology , Bacteria/drug effects , Blindness/prevention & control , Global Health , Humans , Morbidity , Prognosis
6.
Curr Eye Res ; 31(11): 955-65, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17114121

ABSTRACT

Rapid vision loss and explosive inflammation are devastating consequences of Bacillus endophthalmitis that have not been well defined. We therefore analyzed the evolution of intraocular inflammation and loss of retinal architecture and function during experimental Bacillus endophthalmitis. Mice were intravitreally injected with 100 CFU of B. cereus, and eyes were analyzed for bacterial growth, retinal function, architectural changes and retinal cellular stress, inflammatory cytokines, and infiltrating cells. Retinal electrophysiologic and structural changes began as early as 4 to 6 hr postinfection. Significant declines in retinal function paralleled the loss of retinal architecture. Glial fibrillary acidic protein (GFAP) was detected in retina, indicating potential stress. Polymorphonuclear leukocyte (PMN) infiltration into the vitreous began as early as 4 hr postinfection, coinciding with a significant increase in TNF-alpha in the eye. These results indicated that acute inflammation and detrimental architectural and electrophysiologic changes in the retina began earlier than once thought, suggesting that therapeutic intervention should be given at the earliest possible time to avoid vision loss during Bacillus endophthalmitis.


Subject(s)
Bacillaceae Infections/microbiology , Bacillus cereus/pathogenicity , Endophthalmitis/microbiology , Eye Infections, Bacterial/microbiology , Retinal Diseases/microbiology , Acute Disease , Animals , Bacillaceae Infections/pathology , Disease Models, Animal , Endophthalmitis/pathology , Eye Infections, Bacterial/pathology , Flow Cytometry , Glial Fibrillary Acidic Protein/metabolism , Immunohistochemistry , Inflammation/microbiology , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Neutrophils/physiology , Peroxidase/metabolism , Retinal Diseases/metabolism , Retinal Diseases/pathology , Tumor Necrosis Factor-alpha/metabolism
7.
Curr Eye Res ; 31(9): 693-702, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16966141

ABSTRACT

Bacillus causes one of the most rapidly blinding intraocular infections: endophthalmitis. In this study, Bacillus spp. were isolated from ocular infection cases, taxonomically characterized by riboprint analysis, and screened for the presence of putative virulence factors. The ability of these isolates to kill retinal and corneal cells was examined, as were antibiotic susceptibility profiles. The majority of isolates belonged to the B. cereus taxonomic group of microorganisms and were identified as B. cereus (53%) or B. thuringiensis (26%). Toxins were identified in most B. thuringiensis and B. cereus isolates. Most B. cereus and B. thuringiensis killed corneal and retinal cells within 6 h. All isolates were susceptible to most antibiotics tested, with quinolones and vancomycin being the most potent. These findings represent the first report of B. thuringiensis as an important ocular pathogen, demonstrates the potential ocular toxicity of B. cereus and B. thuringiensis isolates, and identifies antibiotics whose efficacy against Bacillus were superior to those used clinically.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacillus/drug effects , Bacillus/metabolism , Bacterial Proteins/metabolism , Virulence Factors/metabolism , Bacillus/isolation & purification , Bacterial Proteins/genetics , Bacterial Typing Techniques , Cells, Cultured , Connective Tissue Cells/microbiology , Cornea/cytology , DNA, Bacterial/analysis , Eye Infections, Bacterial/microbiology , Genotype , Humans , Microbial Sensitivity Tests , Polymerase Chain Reaction , Retina/cytology , Virulence/physiology , Virulence Factors/genetics
8.
Am J Ophthalmol ; 140(3): 363-9, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16084788

ABSTRACT

PURPOSE: Adenoviruses typically demonstrate specific tissue tropisms, as in the association of Ad19 with epidemic keratoconjunctivitis. We sought to determine factors that might influence the apparent tropism of Ad19 for the cornea. DESIGN: Laboratory investigation. METHODS: Adenovirus serotypes Ad2, 5, 9, 10, 11, 13, and 19 were compared for their capacity to replicate in human corneal epithelial cells (HCECs) in culture. Organotypically cultured human corneas were infected with Ad19 or Ad2, and viral titers were compared after 7 days. Replication of both viruses was compared in HCECs cultured on various extracellular matrices. Western blot analysis and immunohistochemistry were applied to human donor corneas and HCECs. RESULTS: One week after infection of HCEC monolayer cultures, Ad2 titers were significantly higher than any of the other viruses tested (P <.05). In organotypic corneal cultures, Ad19 titers were significantly higher than Ad2 (P = .0003). Ad2 replication in HCECs equaled or exceeded that of Ad19 on all extracellular matrices except vitronectin, where Ad2 replication was reduced and Ad19 replication enhanced (P <.0001). Vitronectin was detected by immunohistochemistry within the corneal epithelial basement membranes of human donor corneas. Increased alpha(v) integrin expression and greater tyrosine kinase phosphorylation in HCECs cultured on vitronectin were demonstrated by Western blot analysis. CONCLUSIONS: In vitro, vitronectin enhances growth of Ad19, possibly by up-regulation of receptor alpha(v) integrins and increased activity of tyrosine kinases necessary for adenoviral internalization. We hypothesize that differential tissue tropisms for adenoviruses may derive in part from tissue-specific extracellular matrix expression.


Subject(s)
Adenoviruses, Human/physiology , Epithelium, Corneal/virology , Tropism/physiology , Vitronectin/physiology , Blotting, Western , Cells, Cultured , Electrophoresis, Polyacrylamide Gel , Humans , Immunohistochemistry , Integrin alphaV/metabolism , Phosphorylation , Tyrosine/metabolism , Virus Replication/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...