Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 2.915
1.
BMC Ophthalmol ; 24(1): 214, 2024 May 17.
Article En | MEDLINE | ID: mdl-38760776

BACKGROUND: Endogenous endophthalmitis (EE) is a rare but highly destructive eye emergency secondary to systemic infection. Acute endophthalmitis can lead to irreversible vision impairment or even loss of the whole eye, unless being diagnosed and treated promptly. CASE PRESENTATION: This study reports three typical EE cases of endogenous endophthalmitis secondary to different severe systemic diseases. Patients were recruited from the Department of ophthalmology at Zhongnan hospital of Wuhan University and the Department of ophthalmology at the Second Affiliated Hospital of Fujian Medical University. Patients were followed up for up to 60 days. Among these cases, the eye symptoms is the initial manifestations while secondary to original different special systemic conditions. Patients have been treated under dynamically prompt response undergoing systemic treatment and eye treatment at the same time. Best corrected visual acuity were 20/40, 20/60 and light perception during follow-up evaluation. CONCLUSIONS: Our observation suggest that prompt identification and treatment could save patients' vision from EE.


Endophthalmitis , Eye Infections, Bacterial , Visual Acuity , Humans , Endophthalmitis/diagnosis , Endophthalmitis/microbiology , Male , Female , Middle Aged , Eye Infections, Bacterial/microbiology , Eye Infections, Bacterial/diagnosis , Eye Infections, Bacterial/drug therapy , Visual Acuity/physiology , Aged , Anti-Bacterial Agents/therapeutic use , Adult
2.
Medicina (Kaunas) ; 60(5)2024 Apr 25.
Article En | MEDLINE | ID: mdl-38792883

We present a case of endogenous endophthalmitis with urinary tract infection (UTI) caused by group B Streptococcus (GBS). An 86-year-old female initially presented with ocular pain and sudden visual disturbance of the left eye. The patient did not complain of other symptoms and had no history of recent ocular surgery or trauma. Endogenous endophthalmitis was clinically diagnosed based on ophthalmic examination, history, and lab results showing systemic infection. A few days later, GBS was identified in her aqueous humor, blood, and urine cultures. Intravitreal ceftazidime and vancomycin injections, as well as fortified ceftazidime and vancomycin eye drops, were used immediately after clinical diagnosis. However, the symptoms worsened despite repeated intravitreal injections, so evisceration was performed. Endogenous endophthalmitis caused by GBS is very virulent and may present without evident systemic symptoms. The early recognition of the disease and systemic work up, followed by prompt treatment, is necessary.


Anti-Bacterial Agents , Endophthalmitis , Streptococcal Infections , Streptococcus agalactiae , Urinary Tract Infections , Humans , Female , Urinary Tract Infections/diagnosis , Urinary Tract Infections/drug therapy , Urinary Tract Infections/microbiology , Urinary Tract Infections/complications , Aged, 80 and over , Endophthalmitis/diagnosis , Endophthalmitis/microbiology , Endophthalmitis/drug therapy , Streptococcus agalactiae/isolation & purification , Streptococcal Infections/drug therapy , Streptococcal Infections/diagnosis , Anti-Bacterial Agents/therapeutic use , Vancomycin/therapeutic use , Ceftazidime/therapeutic use , Ceftazidime/administration & dosage
3.
Transl Vis Sci Technol ; 13(5): 14, 2024 May 01.
Article En | MEDLINE | ID: mdl-38767905

Purpose: Extracellular vesicles (EVs) are messenger pigeons of the cells that communicate about cellular microenvironment. In this study, we evaluated the expression of C8α and calpain-2 in EVs from vitreous of patients with bacterial endophthalmitis to assess its utility as a diagnostic marker. Methods: EVs were isolated from vitreous of patients with bacterial endophthalmitis (culture positive and culture negative) and noninfectious control by exosome isolation reagent and characterized, and the levels of C8α and calpain-2 was assessed by enzyme-linked immunosorbent assay in isolated EVs and direct vitreous. The receiver operating characteristic curve was generated to assess the diagnostic performance. Results: Scanning electron microscopy (SEM) and dynamic light scattering (DLS) confirmed the presence of EVs having a diameter (nm) of 275.2 ± 93, 92 ± 22, and 77.28 ± 12 in culture-positive (CP), culture-negative (CN), and control respectively. The expression level (ng/mL) of C8α in the EVs obtained from CP was 144 ± 22 and CN was 31.2 ± 9.8, which was significantly higher (P < 0.01) than control 3.7 ± 2.4. Interestingly, C8α is not expressed directly in the vitreous of CN and controls. Calpain-2 was significantly downregulated (P ≤ 0.0001) in CP (0.94 ± 0.16) and CN (0.70 ± 0.14) than control. The sensitivity and specificity of 1 for C8α and calpain-2 in the EVs implied that its diagnostic accuracy was significant. Conclusions: This study showed that the EV proteins C8α and calpain-2 could be suitable diagnostic markers for endophthalmitis. However, the presence of C8α in the EVs of CN samples but not in direct vitreous promises EVs as the future of diagnostics. Translational Relevance: Expression levels of EV-calpain-2 and EV-C8α could diagnose CN bacterial endophthalmitis.


Biomarkers , Calpain , Endophthalmitis , Extracellular Vesicles , Vitreous Body , Calpain/metabolism , Humans , Vitreous Body/metabolism , Vitreous Body/microbiology , Endophthalmitis/diagnosis , Endophthalmitis/microbiology , Endophthalmitis/metabolism , Endophthalmitis/pathology , Extracellular Vesicles/metabolism , Biomarkers/metabolism , Male , Female , Middle Aged , Enzyme-Linked Immunosorbent Assay , Aged , Eye Infections, Bacterial/microbiology , Eye Infections, Bacterial/diagnosis , Eye Infections, Bacterial/metabolism , Eye Infections, Bacterial/pathology , ROC Curve , Microscopy, Electron, Scanning , Adult
5.
Invest Ophthalmol Vis Sci ; 65(4): 44, 2024 Apr 01.
Article En | MEDLINE | ID: mdl-38687493

Purpose: Fungal endophthalmitis is characterized by chronic inflammation leading to the partial or complete vision loss. Herein, we analyzed the transcriptomic landscape of Aspergillus flavus (A. flavus) endophthalmitis in C57BL/6 mice to understand the host-pathogen interactions. Methods: Endophthalmitis was induced by intravitreal injection of A. flavus spores in C57BL/6 mice and monitored for disease progression up to 72 hours. The enucleated eyeballs were subjected to histopathological analysis and mRNA sequencing using the Illumina Nextseq 2000. Pathway enrichment analysis was performed to further annotate the functions of differentially expressed genes (DEGs) and validation of cytokines was performed in vitreous of patients with fungal endophthalmitis using multiplex ELISA. Results: Transcriptomic landscape of A. flavus endophthalmitis revealed upregulated T-cell receptor signaling, PI3K-AKT, MAPK, NF-κB, JAK-STAT, and NOD like receptor signaling pathways. We observed significant increase in the T-cells during infection especially at 72 hours infection along with elevated expression levels of IL-6, IL-10, IL-12, IL-18, IL-19, IL-23, CCR3, and CCR7. Furthermore, host-immune response associated genes, such as T-cell interacting activating receptor, TNF receptor-associated factor 1, TLR1, TLR9, and bradykinin receptor beta 1, were enriched. Histopathological assessment validated the significant increase in inflammatory cells, especially T-cells at 72 hours post-infection along with increased disruption in the retinal architecture. Additionally, IL-6, IL-8, IL-17, TNF-α, and IL-1ß were also significantly elevated, whereas IL-10 was downregulated in vitreous of patients with Aspergillus endophthalmitis. Conclusions: Regulating T-cell influx could be a potential strategy to modulate the excessive inflammation in the retina and potentially aid in better vision recovery in fungal endophthalmitis.


Adaptive Immunity , Aspergillosis , Aspergillus flavus , Cytokines , Disease Models, Animal , Endophthalmitis , Eye Infections, Fungal , Gene Expression Profiling , Immunity, Innate , Mice, Inbred C57BL , Animals , Aspergillus flavus/genetics , Mice , Eye Infections, Fungal/microbiology , Eye Infections, Fungal/genetics , Eye Infections, Fungal/immunology , Endophthalmitis/microbiology , Endophthalmitis/immunology , Endophthalmitis/genetics , Aspergillosis/microbiology , Aspergillosis/genetics , Aspergillosis/immunology , Adaptive Immunity/genetics , Immunity, Innate/genetics , Cytokines/metabolism , Cytokines/genetics , Transcriptome , Enzyme-Linked Immunosorbent Assay , Vitreous Body/microbiology
6.
Arq Bras Oftalmol ; 87(4): e2023, 2024.
Article En | MEDLINE | ID: mdl-38656025

Endophthalmitis is a severe form of purulent inflammation caused by the infection of the intraocular tissues or fluids. This infection infrequently occurs through endogenous routes, which are often correlated with major risk factors. Escherichia coli, a gram-negative rod, can cause endophthalmitis through hematogenous spread. We here report a 59-year-old man who presented to our service with acute visual impairment in his left eye, preceded by floaters. He was taking sirolimus and azathioprine for a transplanted kidney, had undergone catheterization for bladder atresia, and had a history of recurrent E. coli urinary tract infections. On evaluation, the left eye exhibited visual acuity of hand motion, anterior chamber reaction (3+/4+), and intense vitritis (4+/4+) with white flake clusters, which prevented appropriate retinal evaluation. Pars plana vitrectomy was performed, and the culture yielded E. coli. The present case highlights the importance of identifying the signs and symptoms of infection early so that diagnosis and treatment of endophthalmitis can be promptly initiated.


Endophthalmitis , Escherichia coli Infections , Escherichia coli , Humans , Endophthalmitis/microbiology , Male , Middle Aged , Escherichia coli/isolation & purification , Vitrectomy , Eye Infections, Bacterial/microbiology , Eye Infections, Bacterial/diagnosis , Visual Acuity , Anti-Bacterial Agents/therapeutic use
7.
Am J Ophthalmol ; 262: 97-106, 2024 Jun.
Article En | MEDLINE | ID: mdl-38280676

PURPOSE: To evaluate factors that inform systemic antifungal choices in patients with endogenous fungal endophthalmitis (EFE). DESIGN: Single-institution retrospective case series. METHODS: Charts of EFE patients from 2010 to 2023 were reviewed. Patients treated systemically for EFE with a minimum of 14 days of follow-up were included. Outcome measures included time to improvement in vitritis or chorioretinitis, systemic therapy modification, and need for surgical intervention. RESULTS: A total of 20 eyes of 16 patients were included. Candida species were most common (43.8%), followed by culture-negative EFE (37.5%) and Aspergillus species (18.8%). In all, 90% of eyes had vitritis and/or macula-involving chorioretinitis. The majority of Candida infections (60%) or culture-negative EFE (75%) were treated initially with oral antifungals. Patients with a history of immune compromise, positive fungal culture, or positive Fungitell assay were more likely to be treated with early intravenous (IV) antifungal therapy. Two patients required systemic antifungal therapy modification because of worsening chorioretinitis, in 1 case due to voriconazole-resistant Aspergillosis that demonstrated chorioretinal lesion growth despite intravitreal amphotericin B injections and systemic voriconazole, and in the second case due to worsening chorioretinitis from Candida dubliniensis infection that regressed upon switch from oral to IV fluconazole. CONCLUSIONS: Initial systemic treatment decisions in patients with EFE were driven by systemic culture positivity, systemic symptoms, or comorbidities. Intravitreal antifungal therapy may be insufficient to arrest progression of chorioretinal lesions in some cases. Larger studies are needed to determine whether visible end-organ damage in the form of chorioretinitis may be useful for guiding systemic therapy changes.


Antifungal Agents , Endophthalmitis , Eye Infections, Fungal , Humans , Retrospective Studies , Eye Infections, Fungal/microbiology , Eye Infections, Fungal/drug therapy , Eye Infections, Fungal/diagnosis , Endophthalmitis/microbiology , Endophthalmitis/drug therapy , Endophthalmitis/diagnosis , Antifungal Agents/therapeutic use , Male , Female , Middle Aged , Aged , Adult , Aged, 80 and over , Visual Acuity/physiology , Fungi/isolation & purification , Chorioretinitis/microbiology , Chorioretinitis/drug therapy , Chorioretinitis/diagnosis
8.
Prostaglandins Other Lipid Mediat ; 171: 106806, 2024 Apr.
Article En | MEDLINE | ID: mdl-38185280

Bacterial endophthalmitis is a blinding infectious disease typically acquired during ocular surgery. We previously reported significant alterations in retinal metabolism during Staphylococcus (S) aureus endophthalmitis. However, the changes in retinal lipid composition during endophthalmitis are unknown. Here, using a mouse model of S. aureus endophthalmitis and an untargeted lipidomic approach, we comprehensively analyzed temporal alterations in total lipids and oxylipin in retina. Our data showed a time-dependent increase in the levels of lipid classes, sphingolipids, glycerolipids, sterols, and non-esterified fatty acids, whereas levels of phospholipids decreased. Among lipid subclasses, phosphatidylcholine decreased over time. The oxylipin analysis revealed increased prostaglandin-E2, hydroxyeicosatetraenoic acids, docosahexaenoic acid, eicosapentaenoic acid, and α-linolenic acid. In-vitro studies using mouse bone marrow-derived macrophages showed increased lipid droplets and lipid-peroxide formation in response to S. aureus infection. Collectively, these findings suggest that S. aureus-infection alters the retinal lipid profile, which may contribute to the pathogenesis of bacterial endophthalmitis.


Endophthalmitis , Staphylococcus aureus , Humans , Staphylococcus aureus/physiology , Lipidomics , Oxylipins , Endophthalmitis/microbiology , Endophthalmitis/pathology , Retina/pathology
9.
Front Cell Infect Microbiol ; 13: 1304677, 2023.
Article En | MEDLINE | ID: mdl-38106476

Background: Bacillus cereus (Bc) can cause self-limiting gastrointestinal infections, but when infecting the eye, can cause rapid and irreversible blindness. This study investigated whether clinical ocular and gastrointestinal Bc isolates differed in terms of virulence-related genotypes and endophthalmitis virulence. Methods: Twenty-eight Bc ocular, gastrointestinal, and laboratory reference isolates were evaluated. Hemolysis assays were performed to assess potential differences in hemolytic activity. The presence of twenty Bc virulence-related genes was assessed by PCR. A subset of ocular and gastrointestinal isolates differing in PCR positivity for 5 virulence genes was compared to strain ATCC14579 in an experimental murine model of endophthalmitis. At 8 hours post infection, retinal function was evaluated by electroretinography, and intraocular bacterial concentrations were determined by plate counts. Results: Gastrointestinal Bc isolates were more hemolytic than the Bc ocular isolates and ATCC14579 (p < 0.0001). Bc ocular isolates were more frequently PCR-positive for capK, cytK, hblA, hblC, and plcR compared to the gastrointestinal isolates (p ≤ 0.0002). In the endophthalmitis model, mean A-wave retention did not differ significantly between eyes infected with ATCC14579 and eyes infected with the selected ocular or gastrointestinal isolates (p ≥ 0.3528). Similar results were observed for mean B-wave retention (p ≥ 0.0640). Only one diarrheal isolate showed significantly greater B-wave retention when compared to ATCC14579 (p = 0.0303). No significant differences in mean A-wave (p ≥ 0.1535) or B-wave (p ≥ 0.0727) retention between the selected ocular and gastrointestinal isolates were observed. Intraocular concentrations of ATCC14579 were significantly higher than the selected ocular isolate and 3 of the gastrointestinal isolates (p ≤ 0.0303). Intraocular concentrations of the selected ocular isolate were not significantly different from the gastrointestinal isolates (p ≥ 0.1923). Conclusions: Among the subset of virulence-related genes assessed, 5 were significantly enriched among the ocular isolates compared to gastrointestinal isolates. While hemolytic activity was higher among gastrointestinal isolates, retinal function retention and intraocular growth was not significantly different between the selected ocular and gastrointestinal isolates. These results suggest that Bc strains causing gastrointestinal infections, while differing from ocular isolates in hemolytic activity and virulence-related gene profile, are similarly virulent in endophthalmitis.


Bacillus cereus , Endophthalmitis , Mice , Humans , Animals , Bacillus cereus/genetics , Virulence/genetics , Endophthalmitis/microbiology , Endophthalmitis/pathology , Retina , Genotype
10.
BMC Ophthalmol ; 23(1): 450, 2023 Nov 10.
Article En | MEDLINE | ID: mdl-37950172

BACKGROUND: Endophthalmitis following intravitreal injection is a potentially devastating complication of anti-VEGF injections. Post-injection endophthalmitis due to Enterococcus faecalis is rare, and no previous case of Morganella morganii endophthalmitis after intravitreal injection has been reported. CASE PRESENTATION: We present the first reported case of Morganella morganii and Enterococcus faecalis endophthalmitis after intravitreal injection in an immunocompetent patient in the absence of recent ocular surgery. Our patient presented with hand movement visual acuity one day after anti-VEGF injection and demonstrated no clinical improvement despite repeated intravitreal ceftazidime and vancomycin injections. A decision was made to proceed with early vitrectomy given failure of intravitreal antibiotics. Visual acuity improved to 6/90 at 12 weeks after vitrectomy without any evidence of disease recurrence. CONCLUSIONS: Post-injection endophthalmitis due to concurrent Morganella morganii and Enterococcus faecalis infections can have visually devastating consequences despite repeated empirical and targeted intravitreal antibiotics. Lack of clinical improvement following intravitreal antibiotics should warrant consideration of early vitrectomy. Our experience is a pertinent reminder of the ever-growing threat of uncommon and multi-resistant bacteria that must be considered when treating infections such as post-injection endophthalmitis.


Endophthalmitis , Eye Infections, Bacterial , Morganella morganii , Humans , Enterococcus faecalis , Intravitreal Injections , Eye Infections, Bacterial/diagnosis , Eye Infections, Bacterial/drug therapy , Eye Infections, Bacterial/microbiology , Endophthalmitis/diagnosis , Endophthalmitis/drug therapy , Endophthalmitis/microbiology , Anti-Bacterial Agents/therapeutic use , Vitrectomy/adverse effects , Bacteria , Retrospective Studies
11.
Asia Pac J Ophthalmol (Phila) ; 12(5): 437-443, 2023.
Article En | MEDLINE | ID: mdl-37851560

PURPOSE: An interim analysis of the Endophthalmitis Management Study to examine the outcome of inflammation score (IS)-based treatment and antibiotic susceptibility. DESIGN: A prospective randomized study. PATIENTS AND METHODS: IS was measured on a 0-4 scale from presenting signs in 4 cardinal ocular tissues. The eyes with IS <10 received vitreous tap and intravitreal antibiotics, whereas eyes with IS ≥10 received vitrectomy and intravitreal antibiotics. These eyes were randomized to 2 intravitreal antibiotic combinations: (1) vancomycin and ceftazidime and (2) vancomycin and imipenem. Microbiology workup of undiluted vitreous included microscopy, culture-susceptibility, Sanger, and targeted next-generation sequencing. The clinical and microbiology outcomes were analyzed for advanced (IS = ≥20) and less advanced (IS = <10) endophthalmitis. RESULTS: Interim analysis was performed after the Endophthalmitis Management Study recruited 56.85% (248/436) of patients and completed 54.6% (238/436) of microbiology workup. A 90-day follow-up was completed in 90.8% (168/185) of eligible people. In eyes with IS ≥20, the time to symptoms was shorter (5.8 ± 6.7 vs 8.5 ± 9.1 d; P = 0.015), and the need for additional treatment was higher (95.8% vs 53.1%; P = 0.0267). Good final vision was associated with good presenting vision (r = 0.30) and IS-based treatment decisions (r = 0.170). Microbiology positivity was 55.9%. Eyes with IS <10 had a higher Gram-positive cocci (33.9% vs 4.8%; P = 0.013) infection. Gram-positive cocci were most susceptible to vancomycin (95.7%), and Gram-negative bacilli to colistin (95.7%). CONCLUSIONS: Considering both IS and presenting vision, rather than only one of them, helps in making appropriate management decisions for acute postoperative endophthalmitis.


Endophthalmitis , Eye Infections, Bacterial , Humans , Vancomycin/therapeutic use , Prospective Studies , Eye Infections, Bacterial/diagnosis , Eye Infections, Bacterial/drug therapy , Eye Infections, Bacterial/microbiology , Endophthalmitis/diagnosis , Endophthalmitis/drug therapy , Endophthalmitis/microbiology , Anti-Bacterial Agents/therapeutic use , Postoperative Complications/drug therapy , Vitrectomy , Retrospective Studies
13.
Cell Mol Life Sci ; 80(10): 309, 2023 Sep 28.
Article En | MEDLINE | ID: mdl-37770649

Bacterial endophthalmitis is a severe complication of eye surgeries that can lead to vision loss. Current treatment involves intravitreal antibiotic injections that control bacterial growth but not inflammation. To identify newer therapeutic targets to promote inflammation resolution in endophthalmitis, we recently employed an untargeted metabolomics approach. This led to the discovery that the levels of S-nitroso-L-glutathione (GSNO) were significantly reduced in an experimental murine Staphylococcus aureus (SA) endophthalmitis model. In this study, we tested the hypothesis whether GSNO supplementation via different routes (oral, intravitreal) provides protection during bacterial endophthalmitis. Our results show that prophylactic administration of GSNO via intravitreal injections ameliorated SA endophthalmitis. Therapeutically, oral administration of GSNO was found to be most effective in reducing intraocular inflammation and bacterial burden. Moreover, oral GSNO treatment synergized with intravitreal antibiotic injections in reducing the severity of endophthalmitis. Furthermore, in vitro experiments using cultured human retinal Muller glia and retinal pigment epithelial (RPE) cells showed that GSNO treatment reduced SA-induced inflammatory mediators and cell death. Notably, both in-vivo and ex-vivo data showed that GSNO strengthened the outer blood-retinal barrier during endophthalmitis. Collectively, our study demonstrates GSNO as a potential therapeutic agent for the treatment of intraocular infections due to its dual anti-inflammatory and cytoprotective properties.


Endophthalmitis , Eye Infections, Bacterial , Staphylococcal Infections , Mice , Humans , Animals , Disease Models, Animal , Endophthalmitis/drug therapy , Endophthalmitis/microbiology , Staphylococcal Infections/drug therapy , Inflammation/drug therapy , Staphylococcus aureus , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Glutathione , Eye Infections, Bacterial/drug therapy , Eye Infections, Bacterial/microbiology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Administration, Oral
14.
Ugeskr Laeger ; 185(38)2023 09 18.
Article Da | MEDLINE | ID: mdl-37772652

Intravesical instillation of bacillus Calmette-Guérin (BCG) is used for the maintenance treatment of some types of bladder cancer. Although rare, ocular complications can develop following intravesical BCG treatment. This is a case report of culture-positive Mycobacterium bovis BCG endophthalmitis following intravesical BCG installation. The case highlights a rare complication of BCG installation and the need for an eye examination when patients after BCG installation develop eye symptoms.


Endophthalmitis , Mycobacterium bovis , Urinary Bladder Neoplasms , Humans , BCG Vaccine/therapeutic use , Urinary Bladder Neoplasms/drug therapy , Endophthalmitis/microbiology , Administration, Intravesical
17.
mSphere ; 8(4): e0004423, 2023 08 24.
Article En | MEDLINE | ID: mdl-37273201

Bacteriophage lytic enzymes (i.e., phage lysins) are a trending alternative for general antibiotics to combat growing antimicrobial resistance. Gram-positive Bacillus cereus causes one of the most severe forms of intraocular infection, often resulting in complete vision loss. It is an inherently ß-lactamase-resistant organism that is highly inflammogenic in the eye, and antibiotics are not often beneficial as the sole therapeutic option for these blinding infections. The use of phage lysins as a treatment for B. cereus ocular infection has never been tested or reported. In this study, the phage lysin PlyB was tested in vitro, demonstrating rapid killing of vegetative B. cereus but not its spores. PlyB was also highly group specific and effectively killed the bacteria in various bacterial growth conditions, including ex vivo rabbit vitreous (Vit). Furthermore, PlyB demonstrated no cytotoxic or hemolytic activity toward human retinal cells or erythrocytes and did not trigger innate activation. In in vivo therapeutic experiments, PlyB was effective in killing B. cereus when administered intravitreally in an experimental endophthalmitis model and topically in an experimental keratitis model. In both models of ocular infection, the effective bactericidal property of PlyB prevented pathological damage to ocular tissues. Thus, PlyB was found to be safe and effective in killing B. cereus in the eye, greatly improving an otherwise devastating outcome. Overall, this study demonstrates that PlyB is a promising therapeutic option for B. cereus eye infections.IMPORTANCEEye infections from antibiotic-resistant Bacillus cereus are devastating and can result in blindness with few available treatment options. Bacteriophage lysins are an alternative to conventional antibiotics with the potential to control antibiotic-resistant bacteria. This study demonstrates that a lysin called PlyB can effectively kill B. cereus in two models of B. cereus eye infections, thus treating and preventing the blinding effects of these infections.


Bacillus Phages , Bacillus , Endophthalmitis , Eye Infections, Bacterial , Animals , Humans , Rabbits , Eye Infections, Bacterial/drug therapy , Endophthalmitis/drug therapy , Endophthalmitis/microbiology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use
18.
Arch Microbiol ; 205(6): 236, 2023 May 15.
Article En | MEDLINE | ID: mdl-37183227

Ocular fungal infections annually affect more than one million individuals worldwide. The management of these infections is problematic, mainly due to the limited availability of effective antifungal agents. Thus, ocular infections are increasingly recognized as important causes of morbidity and blindness, especially keratitis and endophthalmitis. Thus, this review aims to demonstrate the importance of fungal eye infections through the description of the main related aspects, with emphasis on the treatment of these infections. For this purpose, a search for scientific articles was conducted in databases, such as Medline, published from 2000 onwards, addressing important aspects involving fungal eye infections. In addition, this work highlighted the limited therapeutic arsenal available and the severity associated with these infections. Thus, highlighting the importance of constantly updating knowledge about these pathologies, as it contributes to agility in choosing the available and most appropriate therapeutic alternatives, aiming at positive and minimally harmful results for that particular patient.


Endophthalmitis , Eye Infections, Fungal , Keratitis , Humans , Eye Infections, Fungal/diagnosis , Eye Infections, Fungal/drug therapy , Eye Infections, Fungal/microbiology , Antifungal Agents/therapeutic use , Keratitis/drug therapy , Keratitis/microbiology , Endophthalmitis/diagnosis , Endophthalmitis/drug therapy , Endophthalmitis/microbiology
19.
Graefes Arch Clin Exp Ophthalmol ; 261(10): 2813-2819, 2023 Oct.
Article En | MEDLINE | ID: mdl-37227476

PURPOSE: Increasing rates of antibiotic resistance in endophthalmitis have been reported. This study examines outcomes of triple therapy with intravitreal vancomycin, ceftazidime, and moxifloxacin for endophthalmitis. METHODS: Retrospective, consecutive series of all patients treated with abovementioned intravitreal antibiotics from January 2009 to June 2021. Percentages of eyes attaining greater than or equal to 20/200 and 20/50 Snellen visual acuities and adverse events were evaluated. RESULTS: 112 eyes met inclusion criteria. 63 of 112 eyes (56%) achieved a visual acuity of 20/200 during follow-up, with 39 (35%) returning to at least 20/50. In subgroup analysis, 23 of 24 (96%) eyes with post-cataract endophthalmitis obtained ≥ 20/200 acuity and 21 of 24 (88%) obtained ≥ 20/50 acuity during follow-up. There were no cases of macular infarction. CONCLUSIONS: Intravitreal moxifloxacin (160 µg/0.1 mL) was well tolerated as an adjunct to vancomycin and ceftazidime for bacterial endophthalmitis. Use of this novel combination offers several theoretical advantages compared to standard therapy with two antibiotics, including expanded gram-negative coverage and potential synergy, and may be particularly valuable in geographies where the local antibiogram supports empiric use. Further study is merited to verify the safety and efficacy profile.


Endophthalmitis , Eye Infections, Bacterial , Humans , Vancomycin/therapeutic use , Ceftazidime/therapeutic use , Moxifloxacin , Retrospective Studies , Vitreous Body/microbiology , Anti-Bacterial Agents , Endophthalmitis/diagnosis , Endophthalmitis/drug therapy , Endophthalmitis/microbiology , Eye Infections, Bacterial/diagnosis , Eye Infections, Bacterial/drug therapy , Eye Infections, Bacterial/microbiology
...