1
|
Zhang Z, Zhao R, Wu X, Ma Y, He Y. Research progress on the correlation between corneal neovascularization and lymphangiogenesis (Review). Mol Med Rep 2025; 31:47. [PMID: 39635819 PMCID: PMC11638739 DOI: 10.3892/mmr.2024.13412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 10/23/2024] [Indexed: 12/07/2024] Open
Abstract
The cornea is a clear connective tissue membrane at the front of the outer layer of the eyeball wall. It plays a crucial role in the refractive system of the eyeball, making it essential to maintain its transparency. Neovascularization and lymphangiogenesis in the cornea significantly impact corneal transparency and immune privilege. The growth of corneal neovascularization (CNV) and corneal lymphangiogenesis (CL) vessels is interconnected yet independent. Currently, there is a substantial amount of clinical and experimental research on CNV and CL vessels. However, due to the relatively recent focus on CL vessel research compared with CNV research, most scholars tend to concentrate on CNV, with few articles offering a comprehensive comparison and discussion of the two processes. The present review emphasizes the similarities and differences between CNV and CL and summarizes recent research progress on their correlation in animal models, growth characteristics, cytokine effects and related diseases.
Collapse
Affiliation(s)
- Zhaochen Zhang
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Rongxuan Zhao
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, Jilin 130041, P.R. China
| | - Xuhui Wu
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Yunkun Ma
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Yuxi He
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| |
Collapse
|
2
|
Bertret C, Knoeri J, Leveziel L, Bourcier T, Brignole-Baudouin F, Merabet L, Bouheraoua N, Borderie VM. Predisposing factors, clinical and microbiological insights of bacterial keratitis: analysis of 354 cases from a leading French academic centre. Br J Ophthalmol 2024; 109:15-20. [PMID: 38925906 DOI: 10.1136/bjo-2024-325261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 05/09/2024] [Indexed: 06/28/2024]
Abstract
AIMS To report an epidemiological update of bacterial keratitis (BK) in a tertiary ophthalmology centre over 20 months compared with a previous study on the same timeframe from 1998 to 1999. METHODS 354 patients with BK documented by microbiological corneal scraping or resolutive under antibiotics treatment from January 2020 to September 2021 were analysed retrospectively. RESULTS One or several risk factors were found in 95.2% of patients: contact lens wear (45.2%), ocular surface disease (25.0%), systemic disease (21.8%), ocular trauma (11.9%) and ocular surgery (8.8%). The positivity rate of corneal scrapings was 82.5%, with 18.2% polybacterial. One hundred seventy-five (59.9%) bacteria were Gram-negative, and 117 (40.1%) were Gram-positive. The most common bacteria were Pseudomonas aeruginosa (32.5%), Moraxella spp (18.1%) and Staphylococcus aureus (8.2%). Final visual acuity (logarithm of the minimum angle of resolution) was associated with age (r=+0.48; p=0.0001), infiltrate size (r=+0.32; p<0.0001), ocular surface disease (r=+0.13; p=0.03), ocular trauma (r=-0.14; p=0.02) and contact lens wear (r=-0.26; p<0.0001). Gram-negative bacteria were responsible for deeper (r=+0.18; p=0.004) and more extensive infiltrates (r=+0.18; p=0.004) in younger patients (r=-0.19; p=0.003). Compared with the previous period, the positivity rate of corneal scrapings and the proportion of Gram-negative bacteria, especially Moraxella spp, increased. All P. aeruginosa and Moraxella spp were sensitive to quinolones, and all S. aureus were sensitive to both quinolones and methicillin. CONCLUSION Contact lens wear remained the leading risk factor. The bacteria distribution was reversed, with a predominance of Gram-negative bacteria and increased Moraxella spp.
Collapse
|
3
|
Bourcier T, Koestel E, Bertret C, Yaïci R, Borderie V, Bouheraoua N. [Bacterial keratitis: Retrospective and prospective 2024]. J Fr Ophtalmol 2024; 47:104335. [PMID: 39454484 DOI: 10.1016/j.jfo.2024.104335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 07/08/2024] [Accepted: 07/10/2024] [Indexed: 10/28/2024]
Abstract
Bacterial keratitis (BK) is an infection of the cornea caused by one or more bacteria. Contact lens wear is the main risk factor. Staphylococcus and Pseudomonas are the most frequently isolated pathogens in developed countries. BK requires a standardized work-up to avoid diagnostic and therapeutic delays that may negatively affect visual prognosis. Corneal signs, the speed at which lesions progress and the presence of risk factors allow the clinician to presume an empirical microbiological diagnosis, but corneal scraping, which allows the isolation and identification of the bacteria involved in the infection, is the only way to confirm the diagnosis. The type of antibiotic treatment depends on the severity of the lesions, the risk factors involved, and the bacteria identified. Corticosteroids have been shown to be effective as adjuvant therapy and may be used under certain well-defined circumstances. Surgical treatment is sometimes necessary.
Collapse
Affiliation(s)
- T Bourcier
- Service d'ophtalmologie, hôpitaux universitaires de Strasbourg, université de Strasbourg, Strasbourg, France; Gepromed, The Medical Hub for Patient Safety, Strasbourg, France.
| | - E Koestel
- Service d'ophtalmologie, hôpitaux universitaires de Strasbourg, université de Strasbourg, Strasbourg, France; Gepromed, The Medical Hub for Patient Safety, Strasbourg, France; IHU ForeSight, Inserm-DGOS CIC 1423, Institut de la vision, Paris, France
| | - C Bertret
- Service d'ophtalmologie 5, Hôpital national de la vision des 1520, Paris, France
| | - R Yaïci
- Service d'ophtalmologie, hôpitaux universitaires de Strasbourg, université de Strasbourg, Strasbourg, France; Gepromed, The Medical Hub for Patient Safety, Strasbourg, France
| | - V Borderie
- Service d'ophtalmologie 5, Hôpital national de la vision des 1520, Paris, France; IHU ForeSight, Inserm-DGOS CIC 1423, Institut de la vision, Paris, France
| | - N Bouheraoua
- Service d'ophtalmologie 5, Hôpital national de la vision des 1520, Paris, France; IHU ForeSight, Inserm-DGOS CIC 1423, Institut de la vision, Paris, France
| |
Collapse
|
4
|
Habibullah S, Swain R, Das M, Bhuyan SK, Mohanty B, Mallick S. Engineered PVA-tamarind gum-based biocomposite for sustained ophthalmic delivery of moxifloxacin: Effect of nanocellulose on physicochemical, mechanoelectrical and permeation kinetics. Int J Biol Macromol 2024; 283:137712. [PMID: 39557255 DOI: 10.1016/j.ijbiomac.2024.137712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 10/29/2024] [Accepted: 11/14/2024] [Indexed: 11/20/2024]
Abstract
Widely used polysaccharide-based films in ophthalmic drug delivery have major limitations of inadequate mechanical strength, poor electrical conductivity, and insufficient ocular drug permeability. Moxifloxacin (MFX) biocomposite film of adequate mechanoelectrical properties was developed for sustained ophthalmic drug delivery. Nanocellulose (NC) incorporated (2.5, 5.0, 7.5, and 10.0 %) PVA-tamarind gum-based moxifloxacin composite was prepared using solvent casting method. The addition of NC improved the mechanical properties of the film, demonstrating its ability to strengthen the structure. Stress relaxation (SR) of the film has been augmented (64.67±7.55 to 73.15±0.34 %) due to increased content of NC (0 to 10 %) respectively. Film containing 5 % NC showed the critical edge of tensile strength (11.9±0.39 MPa), and also the threshold limit of electrical conductivity (4.5*107 Ω). The same film exhibited continued drug release as well as erosion-controlled sustained ocular permeation (pH 7.4) and revealed the highest antibacterial activity (ZOI of disc diffusion, cm) with Pseudomonas aeruginosa (4.63±0.15) and Staphylococcus aureus (4.30±0.26) of MFX (≈224 μg). Notably, incorporating NC produced non-irritating and safe for corneal delivery as confirmed by the Draize model test. Our findings suggested that the NC-containing PVA-tamarind gum-based composite film holds a promising approach for sustained ophthalmic delivery of MFX.
Collapse
Affiliation(s)
- Sk Habibullah
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be) University, Bhubaneswar 751003, Odisha, India
| | - Rakesh Swain
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be) University, Bhubaneswar 751003, Odisha, India
| | - Mouli Das
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be) University, Bhubaneswar 751003, Odisha, India
| | - Sisir Kumar Bhuyan
- Department of Pharmaceutics, Institute of Pharmacy and Technology, Salipur-754202, Cuttack, Odisha, India
| | - Biswaranjan Mohanty
- Department of Pharmaceutics, Institute of Pharmacy and Technology, Salipur-754202, Cuttack, Odisha, India.
| | - Subrata Mallick
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be) University, Bhubaneswar 751003, Odisha, India.
| |
Collapse
|
5
|
Wu KY, Qian SY, Faucher A, Tran SD. Advancements in Hydrogels for Corneal Healing and Tissue Engineering. Gels 2024; 10:662. [PMID: 39451315 PMCID: PMC11507397 DOI: 10.3390/gels10100662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 10/08/2024] [Accepted: 10/14/2024] [Indexed: 10/26/2024] Open
Abstract
Hydrogels have garnered significant attention for their versatile applications across various fields, including biomedical engineering. This review delves into the fundamentals of hydrogels, exploring their definition, properties, and classification. Hydrogels, as three-dimensional networks of crosslinked polymers, possess tunable properties such as biocompatibility, mechanical strength, and hydrophilicity, making them ideal for medical applications. Uniquely, this article offers original insights into the application of hydrogels specifically for corneal tissue engineering, bridging a gap in current research. The review further examines the anatomical and functional complexities of the cornea, highlighting the challenges associated with corneal pathologies and the current reliance on donor corneas for transplantation. Considering the global shortage of donor corneas, this review discusses the potential of hydrogel-based materials in corneal tissue engineering. Emphasis is placed on the synthesis processes, including physical and chemical crosslinking, and the integration of bioactive molecules. Stimuli-responsive hydrogels, which react to environmental triggers, are identified as promising tools for drug delivery and tissue repair. Additionally, clinical applications of hydrogels in corneal pathologies are explored, showcasing their efficacy in various trials. Finally, the review addresses the challenges of regulatory approval and the need for further research to fully realize the potential of hydrogels in corneal tissue engineering, offering a promising outlook for future developments in this field.
Collapse
Affiliation(s)
- Kevin Y. Wu
- Department of Surgery, Division of Ophthalmology, University of Sherbrooke, Sherbrooke, QC J1G 2E8, Canada; (K.Y.W.)
| | - Shu Yu Qian
- Faculty of Medicine, University of Sherbrooke, Sherbrooke, QC J1G 2E8, Canada
| | - Anne Faucher
- Department of Surgery, Division of Ophthalmology, University of Sherbrooke, Sherbrooke, QC J1G 2E8, Canada; (K.Y.W.)
| | - Simon D. Tran
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC H3A 1G1, Canada
| |
Collapse
|
6
|
He X, Zhang Z, Hu M, Lin X, Weng X, Lu J, Fang L, Chen X. Liquiritin Alleviates Inflammation in Lipopolysaccharide-Induced Human Corneal Epithelial Cells. Curr Eye Res 2024; 49:930-941. [PMID: 38767463 DOI: 10.1080/02713683.2024.2353263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 04/09/2024] [Accepted: 05/04/2024] [Indexed: 05/22/2024]
Abstract
PURPOSE This research was designed to elucidate the anti-inflammatory impacts of liquiritin on lipopolysaccharide (LPS)-activated human corneal epithelial cells (HCECs). METHODS The Cell Counting kit-8 (CCK-8) assay was adopted to assess cell viability. The enzyme-linked immunosorbent assay (ELISA) was used to detect the secretion levels of the proinflammatory cytokines IL-6, IL-8, and TNF-α. Transcriptome analysis was conducted to identify the genes that exhibited differential expression between different treatment. The model group included cells treated with LPS (10 µg/mL), the treatment group comprised cells treated with liquiritin (80 µM) and LPS (10 µg/mL), and the control group consisted of untreated cells. To further validate the expression levels of the selected genes, including CSF2, CXCL1, CXCL2, CXCL8, IL1A, IL1B, IL24, IL6, and LTB, quantitative real-time PCR was performed. The expression of proteins related to the Akt/NF-κB signaling pathway was assessed through western blot analysis. NF-κB nuclear translocation was evaluated through immunofluorescence staining. RESULTS The secretion of IL-6, IL-8, and TNF-α in LPS-induced HCECs was significantly downregulated by liquiritin. Based on the transcriptome analysis, the mRNA expression of pro-inflammatory cytokines, namely IL-6, IL-8, IL-1β, IL-24, TNF-α, and IL-1α was overproduced by LPS stimulation, and suppressed after liquiritin treatment. Furthermore, the Western blot results revealed a remarkable reduction in the phosphorylation degrees of NF-κB p65, IκB, and Akt upon treatment with liquiritin. Additionally, immunofluorescence analysis confirmed liquiritin's inhibition of LPS-induced p65 nuclear translocation. CONCLUSIONS Collectively, these findings imply that liquiritin suppresses the expression of proinflammatory cytokines, and the anti-inflammatory impacts of liquiritin may be caused by its repression of the Akt/NF-κB signaling pathway in LPS-induced HCECs. These data indicate that liquiritin could provide a potential therapeutic application for inflammation-associated corneal diseases.
Collapse
Affiliation(s)
- Xian He
- Zhejiang Institute of Medical Device Supervision and Testing, Hangzhou, Zhejiang Province, China
- Key Laboratory of Safety Evaluation of Medical Devices of Zhejiang Province, Hangzhou, Zhejiang Province, China
| | - Ziyang Zhang
- Zhejiang Institute of Medical Device Supervision and Testing, Hangzhou, Zhejiang Province, China
| | - Meili Hu
- Zhejiang Institute of Medical Device Supervision and Testing, Hangzhou, Zhejiang Province, China
| | - Xinyi Lin
- Zhejiang Institute of Medical Device Supervision and Testing, Hangzhou, Zhejiang Province, China
| | - Xu Weng
- Zhejiang Institute of Medical Device Supervision and Testing, Hangzhou, Zhejiang Province, China
| | - Jiajun Lu
- Zhejiang Institute of Medical Device Supervision and Testing, Hangzhou, Zhejiang Province, China
| | - Li Fang
- Zhejiang Institute of Medical Device Supervision and Testing, Hangzhou, Zhejiang Province, China
- Key Laboratory of Safety Evaluation of Medical Devices of Zhejiang Province, Hangzhou, Zhejiang Province, China
| | - Xianhua Chen
- Zhejiang Institute of Medical Device Supervision and Testing, Hangzhou, Zhejiang Province, China
- Key Laboratory of Safety Evaluation of Medical Devices of Zhejiang Province, Hangzhou, Zhejiang Province, China
| |
Collapse
|
7
|
Viberg A, Byström B. Frequency and Outcome of Emergency Penetrating Keratoplasty in Infectious Keratitis in Sweden During the 21st Century. Cornea 2024:00003226-990000000-00634. [PMID: 39046831 DOI: 10.1097/ico.0000000000003638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 06/19/2024] [Indexed: 07/27/2024]
Abstract
PURPOSE To study the frequency over time and outcome of penetrating keratoplasty (PK), "keratoplasty à chaud," in patients with infectious keratitis with 2-year follow-up data. METHODS This register-based study included keratitis cases that had undergone PK in Sweden between 2001 and 2020 and reported to the Swedish Corneal Transplant Register. RESULTS During the study period, 69 eyes were subjected to acute PK due to progressive infectious keratitis. The number increased from 2 annual procedures in the first half of the study period to 5 in the second half (P = 0.01). Preoperative corneal perforation was present in 43.5% (n = 30) of the eyes. Two years after surgery, follow-up data were completed in the register for 53 eyes; of these, 62.3% (n = 33) were considered to have functioning grafts, and 20.8% (n = 11) had experienced a rejection episode. The visual acuity improved from hand motion to counting fingers (P = 0.002), and the proportion of eyes with a visual acuity of ≤1.0 logMAR increased from 5.7% (n = 3) before the surgery to 45.3% (n = 24) at the 2-year follow-up (P < 0.001). CONCLUSIONS The number of active infectious keratitis cases undergoing keratoplasty à chaud increased in Sweden during the 21st century. Most of the cases were successful regarding the structural integrity of the bulb, that is, "had a saved eye" and even a functioning graft 2 years after corneal transplantation. The visual gain was distinct, albeit modest. In cases with severe infectious keratitis, and even a concomitant perforation in the cornea due to the infection, corneal transplantation should continue to be an option.
Collapse
Affiliation(s)
- Andreas Viberg
- Department of Clinical Sciences, Ophthalmology, Umeå University, Umeå, Sweden
| | | |
Collapse
|
8
|
Zheng L, Chen Y, Han Y, Lin J, Fan K, Wang M, Teng T, Yang X, Ke L, Li M, Guo S, Li Z, Wu Y, Li C. Thermosensitive Polyhedral Oligomeric Silsesquioxane Hybrid Hydrogel Enhances the Antibacterial Efficiency of Erythromycin in Bacterial Keratitis. Biomater Res 2024; 28:0033. [PMID: 39040621 PMCID: PMC11260774 DOI: 10.34133/bmr.0033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 04/26/2024] [Indexed: 07/24/2024] Open
Abstract
Bacterial keratitis is a serious ocular infection that can impair vision or even cause blindness. The clinical use of antibiotics is limited due to their low bioavailability and drug resistance. Hence, there is a need to develop a novel drug delivery system for this infectious disease. In this study, erythromycin (EM) was encapsulated into a bifunctional polyhedral oligomeric silsesquioxane (BPOSS) with the backbone of the poly-PEG/PPG urethane (BPEP) hydrogel with the aim of improving the drug efficiency in treating bacterial keratitis. A comprehensive characterization of the BPEP hydrogel was performed, and its biocompatibility was assessed. Furthermore, we carried out the evaluation of the antimicrobial effect of the BPEP-EM hydrogel in S. aureus keratitis using in vivo mouse model. The BPEP hydrogel exhibited self-assembling and thermogelling properties, which assisted the drug loading of drug EM and improved its water solubility. Furthermore, the BPEP hydrogel could effectively bind with mucin on the ocular surface, thereby markedly prolonging the ocular residence time of EM. In vivo testing confirmed that the BPEP-EM hydrogel exerted a potent therapeutic action in the mouse model of bacterial keratitis. In addition, the hydrogel also exhibited an excellent biocompatibility. Our findings demonstrate that the BPEP-EM hydrogel showed a superior therapeutic effect in bacterial keratitis and demonstrated its potential as an ophthalmic formulation.
Collapse
Affiliation(s)
- Lan Zheng
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science & Ocular Surface and Corneal Diseases, Eye Institute & Affiliated Xiamen Eye Center & Affiliated First Hospital, School of Medicine,
Xiamen University, Xiamen 361102, PR China
| | - Ying Chen
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology,
School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, PR China
| | - Yi Han
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science & Ocular Surface and Corneal Diseases, Eye Institute & Affiliated Xiamen Eye Center & Affiliated First Hospital, School of Medicine,
Xiamen University, Xiamen 361102, PR China
- Department of Ophthalmology, The First Affiliated Hospital of University of South China, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
| | - Jingwei Lin
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science & Ocular Surface and Corneal Diseases, Eye Institute & Affiliated Xiamen Eye Center & Affiliated First Hospital, School of Medicine,
Xiamen University, Xiamen 361102, PR China
| | - Kai Fan
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology,
School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, PR China
| | - Mengyuan Wang
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science & Ocular Surface and Corneal Diseases, Eye Institute & Affiliated Xiamen Eye Center & Affiliated First Hospital, School of Medicine,
Xiamen University, Xiamen 361102, PR China
| | - Ting Teng
- Department of Ophthalmology, The First Affiliated Hospital of University of South China, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
| | - Xiuqin Yang
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology,
School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, PR China
| | - Lingjie Ke
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology,
School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, PR China
| | - Muyuan Li
- Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117,Shandong Province, PR China
| | - Shujia Guo
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science & Ocular Surface and Corneal Diseases, Eye Institute & Affiliated Xiamen Eye Center & Affiliated First Hospital, School of Medicine,
Xiamen University, Xiamen 361102, PR China
| | - Zibiao Li
- Huaxia Eye Hospital of Quanzhou, Quanzhou, Fujian 362000, China
| | - Yunlong Wu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology,
School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, PR China
| | - Cheng Li
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science & Ocular Surface and Corneal Diseases, Eye Institute & Affiliated Xiamen Eye Center & Affiliated First Hospital, School of Medicine,
Xiamen University, Xiamen 361102, PR China
- Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117,Shandong Province, PR China
- Huaxia Eye Hospital of Quanzhou, Quanzhou, Fujian 362000, China
| |
Collapse
|
9
|
Xiang J, Zou R, Wang P, Wang X, He X, Liu F, Xu C, Wu A. Nitroreductase-responsive nanoparticles for in situ fluorescence imaging and synergistic antibacterial therapy of bacterial keratitis. Biomaterials 2024; 308:122565. [PMID: 38603823 DOI: 10.1016/j.biomaterials.2024.122565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 03/17/2024] [Accepted: 04/02/2024] [Indexed: 04/13/2024]
Abstract
As bacterial keratitis progresses rapidly, prompt intervention is necessary. Current diagnostic processes are time-consuming and invasive, leading to improper antibiotics for treatment. Therefore, innovative strategies for diagnosing and treating bacterial keratitis are urgently needed. In this study, Cu2-xSe@BSA@NTRP nanoparticles were developed by loading nitroreductase-responsive probes (NTRPs) onto Cu2-xSe@BSA. These nanoparticles exhibited integrated fluorescence imaging and antibacterial capabilities. In vitro and in vivo experiments showed that the nanoparticles produced responsive fluorescence signals in bacteria within 30 min due to an interaction between the released NTRP and bacterial endogenous nitroreductase (NTR). When combined with low-temperature photothermal therapy (PTT), the nanoparticles effectively eliminated E. coli and S. aureus, achieved antibacterial efficacy above 95% and facilitated the re-epithelialization process at the corneal wound site in vivo. Overall, the Cu2-xSe@BSA@NTRP nanoparticles demonstrated potential for rapid, noninvasive in situ diagnosis, treatment, and visualization assessment of therapy effectiveness in bacterial keratitis.
Collapse
Affiliation(s)
- Jing Xiang
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China; Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Cixi Institute of Biomedical Engineering, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
| | - Ruifen Zou
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Cixi Institute of Biomedical Engineering, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China; College of Medical Engineering & the Key Laboratory for Medical Functional Nanomaterials, Jining Medical University, Jining, 272067, China
| | - Pin Wang
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Cixi Institute of Biomedical Engineering, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
| | - Xinfangzi Wang
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Cixi Institute of Biomedical Engineering, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
| | - Xuefei He
- Ningbo No. 2 Hospital, Ningbo, 315000, China
| | - Fang Liu
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China.
| | - Chen Xu
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Cixi Institute of Biomedical Engineering, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China; Advanced Energy Science and Technology Guangdong Laboratory, Huizhou, 516000, China.
| | - Aiguo Wu
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Cixi Institute of Biomedical Engineering, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China; Advanced Energy Science and Technology Guangdong Laboratory, Huizhou, 516000, China.
| |
Collapse
|
10
|
Geng X, Zhang N, Li Z, Zhao M, Zhang H, Li J. Iron-doped nanozymes with spontaneous peroxidase-mimic activity as a promising antibacterial therapy for bacterial keratitis. SMART MEDICINE 2024; 3:e20240004. [PMID: 39188699 PMCID: PMC11236036 DOI: 10.1002/smmd.20240004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 04/23/2024] [Indexed: 08/28/2024]
Abstract
The development of non-antibiotic pharmaceuticals with biocompatible and efficient antibacterial properties is of great significance for the treatment of bacterial keratitis. In this study, we have developed antibacterial iron-doped nanozymes (Fe3+-doped nanozymes, FNEs) with distinguished capacity to fight against bacterial infections. The iron-doped nanozymes are composed of Fe3+ doped zeolitic imidazolate framework-8 (Fe/ZIF-8) and polyethylene imide (PEI), which were functionally coated on the surface of Fe/ZIF-8 and imparted the FNEs with improved water dispersibility and biocompatibility. FNEs possess a significant spontaneous peroxidase-mimic activity without the need for external stimulation, thus elevating cellular reactive oxygen species level by catalyzing local H2O2 at the infection site and resulting in bacteria damaged to death. FNEs eliminated 100% of Staphylococcus aureus within 6 h, and significantly relieved inflammation and bacterial infection levels in mice bacterial keratitis, exhibiting higher bioavailability and a superior therapeutic effect compared to conventional antibiotic eye drops. In addition, the FNEs would not generate drug resistance, suggesting that FNEs have great potential in overcoming infectious diseases caused by antimicrobial resistant bacteria.
Collapse
Affiliation(s)
- Xiwen Geng
- Henan Provincial People's HospitalPeople's Hospital of Zhengzhou UniversityZhengzhouChina
- Pharmaceutical Sciences LaboratoryFaculty of Science and EngineeringÅbo Akademi UniversityTurkuFinland
- Turku Bioscience CentreUniversity of Turku and Åbo Akademi UniversityTurkuFinland
| | - Nan Zhang
- Henan Provincial People's HospitalPeople's Hospital of Zhengzhou UniversityZhengzhouChina
- Henan Eye HospitalZhengzhouChina
| | - Zhanrong Li
- Henan Provincial People's HospitalPeople's Hospital of Zhengzhou UniversityZhengzhouChina
- Henan Eye HospitalZhengzhouChina
| | - Mengyang Zhao
- Henan Provincial People's HospitalPeople's Hospital of Zhengzhou UniversityZhengzhouChina
- Henan Eye HospitalZhengzhouChina
| | - Hongbo Zhang
- Pharmaceutical Sciences LaboratoryFaculty of Science and EngineeringÅbo Akademi UniversityTurkuFinland
- Turku Bioscience CentreUniversity of Turku and Åbo Akademi UniversityTurkuFinland
| | - Jingguo Li
- Henan Provincial People's HospitalPeople's Hospital of Zhengzhou UniversityZhengzhouChina
- Henan Eye HospitalZhengzhouChina
| |
Collapse
|
11
|
Siew L, Ng NBH, Lim CHL. Acute painful red eye in a teenage girl. BMJ 2024; 385:e078598. [PMID: 38782411 DOI: 10.1136/bmj-2023-078598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Affiliation(s)
- Lei Siew
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Nicholas Beng Hui Ng
- National University Hospital, Khoo Teck Puat-National University Children's Medical Institute, Singapore
| | | |
Collapse
|
12
|
Guo RQ, Yang J, Yang YB, Chen YN, Xiao YY, Xiang P, Dong MJ, He MF, Wang YT, Xiao YL, Ke HQ, Liu H. Spectrum and antibiotic sensitivity of bacterial keratitis: a retrospective analysis of eight years in a Tertiary Referral Hospital in Southwest China. Front Cell Infect Microbiol 2024; 14:1363437. [PMID: 38529473 PMCID: PMC10961451 DOI: 10.3389/fcimb.2024.1363437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 02/27/2024] [Indexed: 03/27/2024] Open
Abstract
Purpose The objective of this study was to investigate the epidemiological characteristics, distribution of isolates, prevailing patterns, and antibiotic susceptibility of bacterial keratitis (BK) in a Tertiary Referral Hospital located in Southwest China. Methods A retrospective analysis was conducted on 660 cases of bacterial keratitis occurring between January 2015 and December 2022. The demographic data, predisposing factors, microbial findings, and antibiotic sensitivity profiles were examined. Results Corneal trauma emerged as the most prevalent predisposing factor, accounting for 37.1% of cases. Among these cases, bacterial culture results were positive in 318 cases, 68 species of bacteria were identified. The most common Gram-Positive bacteria isolated overall was the staphylococcus epidermis and the most common Gram-Negative bacteria isolated was Pseudomonas aeruginosa. Methicillin-Resistant Staphylococci accounted for 18.1% of all Gram-Positive bacteria. The detection rate of P. aeruginosa showed an increasing trend over time (Rs=0.738, P=0.037). There was a significant decrease in the percentage of Gram-Negative microorganisms over time (Rs=0.743, P=0.035). The sensitivity of Gram-Positive bacteria to linezolid, vancomycin, tigecycline, quinupristin/dalfopristin, and rifampicin was over 98%. The sensitivity rates of Gram-Negative bacteria to amikacin, meropenem, piperacillin/tazobactam, cefoperazone sodium/sulbactam, ceftazidime, and cefepime were all above 85%. In patients with a history of vegetative trauma, the possibility of BK should be taken into account in addition to the focus on fungal keratitis. Conclusion The microbial composition primarily consists of Gram-Positive cocci and Gram-Negative bacilli. Among the Gram-Positive bacteria, S. epidermidis and Streptococcus pneumoniae are the most frequently encountered, while P. aeruginosa is the predominant Gram-Negative bacteria. To combat Gram-Positive bacteria, vancomycin, linezolid, and rifampicin are considered excellent antimicrobial agents. When targeting Gram-Negative pathogens, third-generation cephalosporins exhibit superior sensitivity compared to first and second-generation counterparts. As an initial empirical treatment for severe cases of bacterial keratitis and those unresponsive to fourth-generation fluoroquinolones in community settings, the combination therapy of vancomycin and tobramycin is a justifiable approach. Bacterial keratitis can be better managed by understanding the local etiology and antibacterial drug susceptibility patterns.
Collapse
Affiliation(s)
- Rui-Qin Guo
- Department of Ophthalmology, Second People’s Hospital of Yunnan Province, The Affiliated Hospital of Yunnan University, The Eye Disease Clinical Medical Research Center of Yunnan Province, The Eye Disease Clinical Medical Center of Yunnan Province, Kunming, China
| | - Ji Yang
- Department of Ophthalmology, Second People’s Hospital of Yunnan Province, The Affiliated Hospital of Yunnan University, The Eye Disease Clinical Medical Research Center of Yunnan Province, The Eye Disease Clinical Medical Center of Yunnan Province, Kunming, China
| | - Ya-Bin Yang
- Department of Ophthalmology, Second People’s Hospital of Yunnan Province, The Affiliated Hospital of Yunnan University, The Eye Disease Clinical Medical Research Center of Yunnan Province, The Eye Disease Clinical Medical Center of Yunnan Province, Kunming, China
| | - Ya-Nan Chen
- Department of Ophthalmology, Second People’s Hospital of Yunnan Province, The Affiliated Hospital of Yunnan University, The Eye Disease Clinical Medical Research Center of Yunnan Province, The Eye Disease Clinical Medical Center of Yunnan Province, Kunming, China
| | - Yu-Yuan Xiao
- Department of Ophthalmology, Second People’s Hospital of Yunnan Province, The Affiliated Hospital of Yunnan University, The Eye Disease Clinical Medical Research Center of Yunnan Province, The Eye Disease Clinical Medical Center of Yunnan Province, Kunming, China
| | - Ping Xiang
- Yunnan Province Innovative Research Team of Environmental Pollution, Food Safety, and Human Health, Institute of Environmental Remediation and Human Health, School of Ecology and Environment, Southwest Forestry University, Kunming, China
| | - Meng-Jie Dong
- Department of Ophthalmology, Second People’s Hospital of Yunnan Province, The Affiliated Hospital of Yunnan University, The Eye Disease Clinical Medical Research Center of Yunnan Province, The Eye Disease Clinical Medical Center of Yunnan Province, Kunming, China
| | - Min-Fang He
- Department of Ophthalmology, Second People’s Hospital of Yunnan Province, The Affiliated Hospital of Yunnan University, The Eye Disease Clinical Medical Research Center of Yunnan Province, The Eye Disease Clinical Medical Center of Yunnan Province, Kunming, China
| | - Yin-Ting Wang
- Department of Ophthalmology, Second People’s Hospital of Yunnan Province, The Affiliated Hospital of Yunnan University, The Eye Disease Clinical Medical Research Center of Yunnan Province, The Eye Disease Clinical Medical Center of Yunnan Province, Kunming, China
| | - Yun-Ling Xiao
- Department of Ophthalmology, Honghe County People’s Hospital, Honghe, China
| | - Hong-Qin Ke
- Department of Ophthalmology, Second People’s Hospital of Yunnan Province, The Affiliated Hospital of Yunnan University, The Eye Disease Clinical Medical Research Center of Yunnan Province, The Eye Disease Clinical Medical Center of Yunnan Province, Kunming, China
| | - Hai Liu
- Department of Ophthalmology, Second People’s Hospital of Yunnan Province, The Affiliated Hospital of Yunnan University, The Eye Disease Clinical Medical Research Center of Yunnan Province, The Eye Disease Clinical Medical Center of Yunnan Province, Kunming, China
| |
Collapse
|
13
|
Kulkarni AS, Thool AR, Daigavane S. Fulminant Eye Infection in a Patient With Nephrotic Syndrome: A Case Report. Cureus 2024; 16:e55842. [PMID: 38590483 PMCID: PMC11000746 DOI: 10.7759/cureus.55842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 03/09/2024] [Indexed: 04/10/2024] Open
Abstract
This case report presents the clinical course of a 53-year-old male farmer with nephrotic syndrome, specifically focal segmental glomerulosclerosis, who developed a fulminant eye infection. While receiving maintenance hemodialysis and immunosuppressive therapy, the patient presented with sudden onset redness, discharge, and decreased vision in his right eye. Initial management with topical antibiotics and steroids failed to halt the progression of the infection, leading to corneal perforation and iris prolapse within a few days. Despite the discontinuation of immunosuppressive medications and initiation of broad-spectrum antimicrobial therapy, the patient's compromised renal function and anaemia precluded surgical intervention. This case underscores the challenges in managing severe ocular infections in immunocompromised patients. It highlights the importance of early recognition, aggressive antimicrobial therapy, and close ophthalmologic monitoring in preventing sight-threatening complications. Despite intensive management, the prognosis for visual recovery in such cases may be poor, emphasizing the need for preventive strategies and careful surveillance in high-risk patient populations.
Collapse
Affiliation(s)
- Aditi S Kulkarni
- Ophthalmology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Archana R Thool
- Ophthalmology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Sachin Daigavane
- Ophthalmology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
14
|
Chen Q, Wang L, Wei Y, Xu X, Guo X, Liang Q. Ferroptosis as a Potential Therapeutic Target for Reducing Inflammation and Corneal Scarring in Bacterial Keratitis. Invest Ophthalmol Vis Sci 2024; 65:29. [PMID: 38381413 PMCID: PMC10893897 DOI: 10.1167/iovs.65.2.29] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 02/03/2024] [Indexed: 02/22/2024] Open
Abstract
Purpose Bacterial keratitis (BK) is a serious ocular infection that can cause severe inflammation and corneal scarring, leading to vision loss. In this study, we aimed to investigate the involvement of ferroptosis in the pathogenesis of BK. Methods Transcriptome analysis was performed to evaluate ferroptosis-related gene expression in human BK corneas. Subsequently, the ferroptosis in mouse models of Pseudomonas aeruginosa keratitis and corneal stromal stem cells (CSSCs) were validated. The mice were treated with levofloxacin (LEV) or levofloxacin combined with ferrostatin-1 (LEV+Fer-1). CSSCs were treated with lipopolysaccharide (LPS) or LPS combined Fer-1. Inflammatory cytokines, α-SMA, and ferroptosis-related regulators were evaluated by RT-qPCR, immunostaining, and Western blot. Iron and reactive oxygen species (ROS) were measured. Results Transcriptome analysis revealed significant alterations in ferroptosis-related genes in human BK corneas. In the BK mouse models, the group treated with LEV+Fer-1 exhibited reduced inflammatory cytokines (MPO, TNF-α, and IFN-γ), decreased corneal scarring and α-SMA expression, and lower Fe3+ compared to the BK and LEV groups. Notably, the LEV+Fer-1 group showed elevated GPX4 and SLC7A11 in contrast to the BK and LEV group. In vitro, Fer-1 treatment effectively restored the alterations of ROS, Fe2+, GPX4, and SLC7A11 induced by LPS in CSSCs. Conclusions Ferroptosis plays a crucial role in the pathogenesis of BK. The inhibition of ferroptosis holds promise for mitigating inflammation, reducing corneal scarring, and ultimately enhancing the prognosis of BK. Consequently, this study provides a potential target for innovative therapeutic strategies for BK, which holds immense potential to transform the treatment of BK.
Collapse
Affiliation(s)
- Qiankun Chen
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing, China
| | - Leying Wang
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing, China
| | - Yuan Wei
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing, China
| | - Xizhan Xu
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing, China
| | - Xiaoyan Guo
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing, China
| | - Qingfeng Liang
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing, China
| |
Collapse
|
15
|
Malani M, Thodikayil AT, Saha S, Nirmal J. Carboxylated nanofibrillated cellulose empowers moxifloxacin to overcome Staphylococcus aureus biofilm in bacterial keratitis. Carbohydr Polym 2024; 324:121558. [PMID: 37985120 DOI: 10.1016/j.carbpol.2023.121558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 10/21/2023] [Accepted: 11/02/2023] [Indexed: 11/22/2023]
Abstract
Bacterial keratitis is one of the vision-threatening ocular diseases that is increasing at an alarming rate due to antimicrobial resistance. One of the primary causes of antimicrobial resistance could be biofilm formation, which alters the mechanism and physiology of the microorganisms. Even a potent drug fails to inhibit biofilm due to the extracellular polysaccharide matrix surrounding the bacteria, inhibiting the permeation of drugs. Therefore, we aimed to develop carboxylated nanocellulose fibers loaded with moxifloxacin (Mox-cNFC) as a novel drug delivery system to treat bacterial corneal infection. Nanocellulose fibers were fabricated using a two-step method involving citric acid hydrolysis followed by TEMPO oxidation to introduce carboxylated groups (1.12 mmol/g). The Mox-cNFC particles showed controlled drug release till 40 h through diffusion. In vitro biofilm inhibition studies showed the particle's ability to disrupt the biofilm matrix and enhance the drug penetration to achieve optimal concentrations that inhibit the persister cells (without increasing minimum inhibitory concentration), thereby reducing the bacterial drug-resistant property. In vivo studies revealed the therapeutic potential of Mox-cNFC to treat Staphylococcus aureus-induced bacterial keratitis with once-a-day treatment, unlike neat moxifloxacin. Mox-cNFC could improve patient compliance by reducing the frequency of instillation and a controlled drug release to prevent toxicity.
Collapse
Affiliation(s)
- Manisha Malani
- Translational Pharmaceutics Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad Campus, Hyderabad 500078, Telangana, India
| | | | - Sampa Saha
- Department of Materials Science and Engineering, Indian Institute of Technology Delhi, New Delhi 110016, India.
| | - Jayabalan Nirmal
- Translational Pharmaceutics Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad Campus, Hyderabad 500078, Telangana, India.
| |
Collapse
|
16
|
Lapp T, Kammrath Betancor P, Schlunck G, Auw-Hädrich C, Maier P, Lange C, Reinhard T, Wolf J. Transcriptional profiling specifies the pathogen-specific human host response to infectious keratitis. Front Cell Infect Microbiol 2024; 13:1285676. [PMID: 38274739 PMCID: PMC10808294 DOI: 10.3389/fcimb.2023.1285676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 12/15/2023] [Indexed: 01/27/2024] Open
Abstract
Purpose Corneal infections are a leading cause of visual impairment and blindness worldwide. Here we applied high-resolution transcriptomic profiling to assess the general and pathogen-specific molecular and cellular mechanisms during human corneal infection. Methods Clinical diagnoses of herpes simplex virus (HSV) (n=5) and bacterial/fungal (n=5) keratitis were confirmed by histology. Healthy corneas (n=7) and keratoconus (n=4) samples served as controls. Formalin-fixed, paraffin-embedded (FFPE) human corneal specimens were analyzed using the 3' RNA sequencing method Massive Analysis of cDNA Ends (MACE RNA-seq). The cellular host response was investigated using comprehensive bioinformatic deconvolution (xCell and CYBERSORTx) analyses and by integration with published single cell RNA-seq data of the human cornea. Results Our analysis identified 216 and 561 genes, that were specifically overexpressed in viral or bacterial/fungal keratitis, respectively, and allowed to distinguish the two etiologies. The virus-specific host response was driven by adaptive immunity and associated molecular signaling pathways, whereas the bacterial/fungal-specific host response mainly involved innate immunity signaling pathways and cell types. We identified several genes and pathways involved in the host response to infectious keratitis, including CXCL9, CXCR3, and MMP9 for viral, and S100A8/A9, MMP9, and the IL17 pathway for bacterial/fungal keratitis. Conclusions High-resolution molecular profiling provides new insights into the human corneal host response to viral and bacterial/fungal infection. Pathogen-specific molecular profiles may provide the foundation for novel diagnostic biomarker and therapeutic approaches that target inflammation-induced damage to corneal host cells with the goal to improve the outcome of infectious keratitis.
Collapse
Affiliation(s)
- Thabo Lapp
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
- Ophtha-Lab, Department of Ophthalmology, St. Franziskus Hospital, Münster, Germany
| | - Paola Kammrath Betancor
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Günther Schlunck
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Claudia Auw-Hädrich
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Philip Maier
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Clemens Lange
- Ophtha-Lab, Department of Ophthalmology, St. Franziskus Hospital, Münster, Germany
| | - Thomas Reinhard
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Julian Wolf
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
- Omics Laboratory, Stanford University, Palo Alto, CA, United States
- Department of Ophthalmology, Byers Eye Institute, Stanford University, Palo Alto, CA, United States
| |
Collapse
|
17
|
Agarwal S, Srinivasan B, Iyer G, Pandey S, Agarwal M, Dhiman R, Surya J, Anand AR. Depth, size of infiltrate, and the microbe - The trio that prognosticates the outcome of infective keratitis. Indian J Ophthalmol 2024; 72:44-50. [PMID: 38131568 PMCID: PMC10841783 DOI: 10.4103/ijo.ijo_1022_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 08/02/2023] [Accepted: 08/23/2023] [Indexed: 12/23/2023] Open
Abstract
PURPOSE To analyze the influence of infiltrate size, depth, and organism on the outcome of microbial keratitis. DESIGN Retrospective comparative study. METHODS Medical records of patients with infective keratitis, who reported from January 2015 to December 2019 to a tertiary eye care center, were analyzed. Size and depth of ulcer at presentation were the factors used to group patients, and the influence on the outcome of the organism causing it was analyzed. Grouping was as follows: group A: ulcer size <6 mm/anterior to midstromal infiltrate, group B: ulcer < 6 mm/full-thickness infiltrate, group C: ulcer >6 mm/anterior to midstromal infiltrate, group D: ulcer > 6 mm/full-thickness infiltrate. Patients with viral keratitis or unidentified organism were excluded. Response to treatment and best-corrected visual acuity (BCVA) at the final follow-up were the outcome measures. RESULTS In the study, 1117/6276 patients were included, with 60.8% patients in group A. A significant improvement in visual acuity was noted in groups A/B compared to groups C/D. Group A had the best response to medical management, irrespective of the organism. Higher risk for surgery was noted in group C compared to group B, with group A as the reference. Overall resolution with medical treatment was noted in 70% miscellaneous keratitis, 64.8% bacterial keratitis, 64.3% mixed keratitis, 62.5% acanthamoeba keratitis, 52.6% fungal keratitis, and 12.1% Pythium keratitis. Bacteria and acanthamoeba responded better to medical management than fungal keratitis, whereas Pythium had the highest risk for surgery. CONCLUSION An interplay between virulence of the organism along with depth and size of the infiltrate determines the outcome of microbial keratitis.
Collapse
Affiliation(s)
- Shweta Agarwal
- CJ Shah Cornea Services, Medical Research Foundation, Sankara Nethralaya, 18, College Road, Chennai, Tamil Nadu, India
| | - Bhaskar Srinivasan
- CJ Shah Cornea Services, Medical Research Foundation, Sankara Nethralaya, 18, College Road, Chennai, Tamil Nadu, India
| | - Geetha Iyer
- CJ Shah Cornea Services, Medical Research Foundation, Sankara Nethralaya, 18, College Road, Chennai, Tamil Nadu, India
| | - Sunita Pandey
- CJ Shah Cornea Services, Medical Research Foundation, Sankara Nethralaya, 18, College Road, Chennai, Tamil Nadu, India
| | - Manokamna Agarwal
- CJ Shah Cornea Services, Medical Research Foundation, Sankara Nethralaya, 18, College Road, Chennai, Tamil Nadu, India
| | - Richa Dhiman
- CJ Shah Cornea Services, Medical Research Foundation, Sankara Nethralaya, 18, College Road, Chennai, Tamil Nadu, India
| | - Janani Surya
- Vision Research Foundation, Sankara Nethralaya, Chennai, Tamil Nadu, India
| | - Appakkudal R Anand
- L and T Microbiology Research Centre, Medical Research Foundation, Sankara Nethralaya, Chennai, Tamil Nadu, India
| | | |
Collapse
|
18
|
Shen X, Huang C, Bai J, Wen J. Targeted Bacterial Keratitis Treatment with Polyethylene Glycol-Dithiothreitol-Boric Acid Hydrogel and Gatifloxacin. Curr Drug Deliv 2024; 21:1548-1558. [PMID: 38425110 DOI: 10.2174/0115672018279105240226050253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 01/18/2024] [Accepted: 02/13/2024] [Indexed: 03/02/2024]
Abstract
INTRODUCTION/OBJECTIVE To prolong the ocular residence time of gatifloxacin and enhance its efficacy against bacterial keratitis, this study developed a velocity-controlled polyethylene glycol-dithiothreitol-boric acid (PDB) hydrogel loaded with gatifloxacin. METHODS First, the basic properties of the synthesized PDB hydrogel and the gatifloxacin-loaded PDB hydrogel were assessed. Secondly, the in vitro degradation rate of the drug-loaded PDB was measured in a simulated body fluid environment with pH 7.4/5.5. The release behavior of the drug-loaded PDB was studied using a dialysis method with PBS solution of pH 7.4/5.5 as the release medium. Finally, a mouse model of bacterial keratitis was established, and tissue morphology was observed using hematoxylin-eosin staining. Additionally, mouse tear fluid was extracted to observe the antibacterial effect of the gatifloxacin-loaded PDB hydrogel. RESULTS The results showed that the PDB hydrogel had a particle size of 124.9 nm and a zeta potential of -23.3 mV, with good porosity, thermosensitivity, viscosity distribution, rheological properties, and high cell compatibility. The encapsulation of gatifloxacin did not alter the physical properties of the PDB hydrogel and maintained appropriate swelling and stability, with a high drug release rate in acidic conditions. Furthermore, animal experiments demonstrated that the gatifloxacin- loaded PDB hydrogel exhibited superior therapeutic effects compared to gatifloxacin eye drops and displayed strong antibacterial capabilities against bacterial keratitis. CONCLUSION This study successfully synthesized PDB hydrogel and developed a gatifloxacin drug release system. The hydrogel exhibited good thermosensitivity, pH responsiveness, stability, and excellent biocompatibility, which can enhance drug retention, utilization, and therapeutic effects on the ocular surface.
Collapse
Affiliation(s)
- Xiao Shen
- Department of Ophthalmology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang, China
| | - Chunlian Huang
- Department of Ophthalmology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang, China
| | - Jianhai Bai
- Department of Ophthalmology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang, China
| | - Jing Wen
- Department of Ophthalmology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang, China
| |
Collapse
|
19
|
Alzahrani M, Kamal YF, Akram MA. Tacrolimus Ointment in Periorbital Atopic Dermatitis. Cureus 2024; 16:e53055. [PMID: 38410340 PMCID: PMC10896248 DOI: 10.7759/cureus.53055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/27/2024] [Indexed: 02/28/2024] Open
Abstract
Periorbital atopic dermatitis (AD) is a common sign in ophthalmological practice and usually has a persistent and relapsing course. Treatment with topical corticosteroids has various side effects associated with their usage. Tacrolimus topical ointment has unique immunomodulatory properties that decrease skin inflammation and pruritus in AD. In this case series, we present a prospective case series of five patients (three males and two females) who received topical application of tacrolimus ointment 0.1-0.03% in the periorbital area twice daily for one to four weeks. The pre- and post-treatment images of all patients were recorded to compare the effects of the treatment. The cases were selected from patients attending the outpatient clinics of East Jeddah Hospital, Saudi Arabia. All patients were suffering from AD. Patients underwent a clinical assessment by tactile inspection (location, size, color, and surface condition) in the first week, secondweek, third month, and first year. We may conclude from this study that tacrolimus showed promising outcomes and is safe and effective for the treatment of flares or resistant periorbital AD in both adults and children.
Collapse
Affiliation(s)
| | - Yumna F Kamal
- Medicine and Surgery, King Abdulaziz University, Jeddah, SAU
| | | |
Collapse
|
20
|
Yu Q, Cai Q, Liang W, Zhong K, Liu J, Li H, Chen Y, Li H, Fang S, Zhong R, Liu S, Lin S. Design of phenothiazine-based cationic amphiphilic derivatives incorporating arginine residues: Potential membrane-active broad-spectrum antimicrobials combating pathogenic bacteria in vitro and in vivo. Eur J Med Chem 2023; 260:115733. [PMID: 37643545 DOI: 10.1016/j.ejmech.2023.115733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 08/14/2023] [Accepted: 08/15/2023] [Indexed: 08/31/2023]
Abstract
Multidrug-resistant bacteria infections pose an increasingly serious threat to human health, and the development of antimicrobials is far from meeting the clinical demand. It is urgent to discover and develop novel antibiotics to combat bacterial resistance. Currently, the development of membrane active antimicrobial agents is an attractive strategy to cope with antimicrobial resistance issues. In this study, the synthesis and biological evaluation of cationic amphiphilic phenothiazine-based derivatives were reported. Among them, the most promising compound 30 bearing a n-heptyl group and two arginine residues displayed potent bactericidal activity against both Gram-positive (MICs = 1.56 μg/mL) and Gram-negative bacteria (MICs = 3.125-6.25 μg/mL). Compound 30 showed low hemolysis activity (HC50 = 281.4 ± 1.6 μg/mL) and low cytotoxicity (CC50 > 50 μg/mL) toward mammalian cells, as well as excellent salt resistance. Compound 30 rapidly killed bacteria by acting on the bacterial cell membrane and appeared less prone to resistance. Importantly, compound 30 showed potent in vivo efficacy in a murine model of bacterial keratitis. Hence, the results suggested compound 30 has a promising prospect as a broad-spectrum antibacterial agent for the treatment of drug-resistant bacterial infections.
Collapse
Affiliation(s)
- Qian Yu
- The Fifth Affiliated Hospital & Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Qiongna Cai
- The Fifth Affiliated Hospital & Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Wanxin Liang
- The Fifth Affiliated Hospital & Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Kewen Zhong
- The Fifth Affiliated Hospital & Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Jiayong Liu
- The Fifth Affiliated Hospital & Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Haizhou Li
- The Fifth Affiliated Hospital & Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Yongzhi Chen
- The Fifth Affiliated Hospital & Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Hongxia Li
- The Fifth Affiliated Hospital & Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Shanfang Fang
- The Fifth Affiliated Hospital & Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Rongcui Zhong
- The Fifth Affiliated Hospital & Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Shouping Liu
- The Fifth Affiliated Hospital & Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, China.
| | - Shuimu Lin
- The Fifth Affiliated Hospital & Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, China.
| |
Collapse
|
21
|
Huang X, Li L, Chen Z, Yu H, You X, Kong N, Tao W, Zhou X, Huang J. Nanomedicine for the Detection and Treatment of Ocular Bacterial Infections. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2302431. [PMID: 37231939 DOI: 10.1002/adma.202302431] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/15/2023] [Indexed: 05/27/2023]
Abstract
Ocular bacterial infection is a prevalent cause of blindness worldwide, with substantial consequences for normal human life. Traditional treatments for ocular bacterial infections areless effective, necessitating the development of novel techniques to enable accurate diagnosis, precise drug delivery, and effective treatment alternatives. With the rapid advancement of nanoscience and biomedicine, increasing emphasis has been placed on multifunctional nanosystems to overcome the challenges posed by ocular bacterial infections. Given the advantages of nanotechnology in the biomedical industry, it can be utilized to diagnose ocular bacterial infections, administer medications, and treat them. In this review, the recent advancements in nanosystems for the detection and treatment of ocular bacterial infections are discussed; this includes the latest application scenarios of nanomaterials for ocular bacterial infections, in addition to the impact of their essential characteristics on bioavailability, tissue permeability, and inflammatory microenvironment. Through an in-depth investigation into the effect of sophisticated ocular barriers, antibacterial drug formulations, and ocular metabolism on drug delivery systems, this review highlights the challenges faced by ophthalmic medicine and encourages basic research and future clinical transformation based on ophthalmic antibacterial nanomedicine.
Collapse
Affiliation(s)
- Xiaomin Huang
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University; Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, 200030, China
- Shanghai Research Center of Ophthalmology and Optometry, Shanghai, 200030, China
- Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Luoyuan Li
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University; Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, 200030, China
- Shanghai Research Center of Ophthalmology and Optometry, Shanghai, 200030, China
- The Eighth Affiliated Hospital Sun Yat-sen University, Shenzhen, Guangdong, 518033, P. R. China
| | - Zhongxing Chen
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University; Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, 200030, China
- Shanghai Research Center of Ophthalmology and Optometry, Shanghai, 200030, China
| | - Haoyu Yu
- The Eighth Affiliated Hospital Sun Yat-sen University, Shenzhen, Guangdong, 518033, P. R. China
| | - Xinru You
- Center for Nanomedicine and Department of Anesthesiology Brigham and Women's Hospital Harvard Medical School, Boston, MA, 02115, USA
| | - Na Kong
- Center for Nanomedicine and Department of Anesthesiology Brigham and Women's Hospital Harvard Medical School, Boston, MA, 02115, USA
| | - Wei Tao
- Center for Nanomedicine and Department of Anesthesiology Brigham and Women's Hospital Harvard Medical School, Boston, MA, 02115, USA
| | - Xingtao Zhou
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University; Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, 200030, China
- Shanghai Research Center of Ophthalmology and Optometry, Shanghai, 200030, China
| | - Jinhai Huang
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University; Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, 200030, China
- Shanghai Research Center of Ophthalmology and Optometry, Shanghai, 200030, China
| |
Collapse
|
22
|
Shanks RMQ, Atta S, Stella NA, Sundar-Raj CV, Romanowski JE, Grewal AS, Shanks HQ, Mumper SM, Dhaliwal DK, Mammen A, Callaghan JD, Calvario RC, Romanowski EG, Kowalski RP, Zegans ME, Jhanji V. A rise in the frequency of lasR mutant Pseudomonas aeruginosa among keratitis isolates between 1993 and 2021. Front Cell Infect Microbiol 2023; 13:1286842. [PMID: 38029269 PMCID: PMC10651084 DOI: 10.3389/fcimb.2023.1286842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction Pseudomonas aeruginosa causes vision threatening keratitis. The LasR transcription factor regulates virulence factors in response to the quorum sensing molecule N-3-oxo-dodecanoyl-L-homoserine lactone. P. aeruginosa isolates with lasR mutations are characterized by an iridescent high sheen phenotype caused by a build-up of 2-heptyl-4-quinolone. A previous study demonstrated 22% (n=101) of P. aeruginosa keratitis isolates from India between 2010 and 2016 were sheen positive lasR mutants, and the sheen phenotype correlated with worse clinical outcomes for patients. In this study, a longitudinal collection of P. aeruginosa keratitis isolates from Eastern North America were screened for lasR mutations by the sheen phenotype and sequencing of the lasR gene. Methods Keratitis isolates (n=399) were classified by sheen phenotype. The lasR gene was cloned from a subset of isolates, sequenced, and tested for loss of function or dominant-negative status based on an azocasein protease assay. A retrospective chart review compared outcomes of keratitis patients infected by sheen positive and negative isolates. Results A significant increase in sheen positive isolates was observed between 1993 and 2021. Extracellular protease activity was reduced among the sheen positive isolates and a defined lasR mutant. Cloned lasR alleles from the sheen positive isolates were loss of function or dominant negative and differed in sequence from previously reported ocular lasR mutant alleles. Retrospective analysis of patient information suggested significantly better visual outcomes for patients infected by sheen positive isolates. Discussion These results indicate an increase in lasR mutations among keratitis isolates in the United States and suggest that endemic lasR mutants can cause keratitis.
Collapse
Affiliation(s)
- Robert M. Q. Shanks
- Charles T. Campbell Laboratory of Ophthalmic Microbiology, Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Sarah Atta
- Charles T. Campbell Laboratory of Ophthalmic Microbiology, Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Nicholas A. Stella
- Charles T. Campbell Laboratory of Ophthalmic Microbiology, Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Chollapadi V. Sundar-Raj
- Charles T. Campbell Laboratory of Ophthalmic Microbiology, Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - John E. Romanowski
- Charles T. Campbell Laboratory of Ophthalmic Microbiology, Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Arman S. Grewal
- Charles T. Campbell Laboratory of Ophthalmic Microbiology, Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Hazel Q. Shanks
- Charles T. Campbell Laboratory of Ophthalmic Microbiology, Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Sonya M. Mumper
- Charles T. Campbell Laboratory of Ophthalmic Microbiology, Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Deepinder K. Dhaliwal
- Charles T. Campbell Laboratory of Ophthalmic Microbiology, Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Alex Mammen
- Charles T. Campbell Laboratory of Ophthalmic Microbiology, Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Jake D. Callaghan
- Charles T. Campbell Laboratory of Ophthalmic Microbiology, Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Rachel C. Calvario
- Charles T. Campbell Laboratory of Ophthalmic Microbiology, Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Eric G. Romanowski
- Charles T. Campbell Laboratory of Ophthalmic Microbiology, Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Regis P. Kowalski
- Charles T. Campbell Laboratory of Ophthalmic Microbiology, Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Michael E. Zegans
- Department of Surgery, Geisel School of Medicine at Dartmouth, Hanover, NH, United States
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, United States
| | - Vishal Jhanji
- Charles T. Campbell Laboratory of Ophthalmic Microbiology, Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
23
|
García-López C, Rodríguez-Calvo-de-Mora M, Borroni D, Sánchez-González JM, Romano V, Rocha-de-Lossada C. The role of matrix metalloproteinases in infectious corneal ulcers. Surv Ophthalmol 2023; 68:929-939. [PMID: 37352980 DOI: 10.1016/j.survophthal.2023.06.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 06/06/2023] [Accepted: 06/19/2023] [Indexed: 06/25/2023]
Abstract
During infectious keratitis, the production of collagenolytic and inflammatory substances, along with increased corneal matrix metalloproteinase (MMP) activity, induces the degradation of corneal collagen and may cause postkeratitis complications, such as opacity, thinning, and corneal perforation. MMPs, especially MMP-2 and MMP-9, are overexpressed in infectious keratitis and sustained over time by inflammatory and nonmicrobial mechanisms. The high MMP levels are correlated with excessive corneal destruction in bacterial, herpetic, fungal, and acanthamoeba infections. Nonspecific treatments, such as tetracyclines, particularly doxycycline, or corticosteroids, are used as adjuvants to antimicrobials to alleviate the disproportionate degradation and inflammation of the corneal layers caused by corneal MMPs and decrease the recruitment and infiltration of inflammatory cells. Treatments showing inhibition of specific MMPs (Galardin, ZHAWOC7726), interfering with pro-MMP activation (EDTA, ascorbic acid), or showing anticytokine effect (epigallocatechin-2-gallate, TRAM-34) have been reported. Other treatments show a direct action over corneal collagen structure such as corneal cross-linking or have been associated with reduction of MMP levels such as amniotic membrane grafting. Although the use of these drugs has been shown in studies to be effective in controlling inflammation, especially in experimental ones, robust studies are still needed based on randomized and randomized clinical trials to demonstrate their potential effect as adjuvants in the management of infectious keratitis.
Collapse
Affiliation(s)
- Celia García-López
- Department of Ophthalmology, Hospital Universitario Virgen de las Nieves, Granada, Spain
| | - Marina Rodríguez-Calvo-de-Mora
- Department of Ophthalmology, Hospital Regional Universitario de Málaga, Málaga, Spain; Department of Ophthalmology (Qvision), Vithas Almería, Almería, Spain; Department of Ophthalmology, VITHAS Málaga, Málaga, Spain
| | - Davide Borroni
- Department of Doctoral Studies, Riga Stradins University, Riga, Latvia; Cornea Research Unit, ADVALIA Vision, Milan, Italy
| | | | - Vito Romano
- Eye Unit, ASST Spedali Civili di Brescia, Brescia, Italy; Eye Unit, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia, Brescia, Italy; Department of Eye and Vision Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Carlos Rocha-de-Lossada
- Department of Ophthalmology, Hospital Regional Universitario de Málaga, Málaga, Spain; Department of Ophthalmology (Qvision), Vithas Almería, Almería, Spain; Department of Ophthalmology, VITHAS Málaga, Málaga, Spain; Department of Surgery, Ophthalmology Area, University of Seville, Seville, Spain
| |
Collapse
|
24
|
Wang M, Li Y, Wang H, Li M, Wang X, Liu R, Zhang D, Xu W. Corneal regeneration strategies: From stem cell therapy to tissue engineered stem cell scaffolds. Biomed Pharmacother 2023; 165:115206. [PMID: 37494785 DOI: 10.1016/j.biopha.2023.115206] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 07/12/2023] [Accepted: 07/18/2023] [Indexed: 07/28/2023] Open
Abstract
Corneal epithelial defects and excessive wound healing might lead to severe complications. As stem cells can self-renew infinitely, they are a promising solution for regenerating the corneal epithelium and treating severe corneal epithelial injury. The chemical and biophysical properties of biological scaffolds, such as the amniotic membrane, fibrin, and hydrogels, can provide the necessary signals for stem cell proliferation and differentiation. Multiple researchers have conducted investigations on these scaffolds and evaluated them as potential therapeutic interventions for corneal disorders. These studies have identified various inherent benefits and drawbacks associated with these scaffolds. In this study, we provided a comprehensive overview of the history and use of various stem cells in corneal repair. We mainly discussed biological scaffolds that are used in stem cell transplantation and innovative materials that are under investigation.
Collapse
Affiliation(s)
- Mengyuan Wang
- Institute of Regenerative Medicine and Laboratory Technology Innovation, Qingdao University, Qingdao, Shandong 266071, PR China
| | - Ying Li
- Institute of Regenerative Medicine and Laboratory Technology Innovation, Qingdao University, Qingdao, Shandong 266071, PR China
| | - Hongqiao Wang
- Blood Purification Department, Qingdao Hospital of Traditional Chinese Medicine, Qingdao Hiser Hospital, Qingdao, Shandong 266071, PR China
| | - Meng Li
- Institute of Regenerative Medicine and Laboratory Technology Innovation, Qingdao University, Qingdao, Shandong 266071, PR China
| | - Xiaomin Wang
- Institute of Regenerative Medicine and Laboratory Technology Innovation, Qingdao University, Qingdao, Shandong 266071, PR China
| | - Rongzhen Liu
- Institute of Regenerative Medicine and Laboratory Technology Innovation, Qingdao University, Qingdao, Shandong 266071, PR China
| | - Daijun Zhang
- Medical College of Qingdao University, Qingdao, Shandong 266071, PR China.
| | - Wenhua Xu
- Institute of Regenerative Medicine and Laboratory Technology Innovation, Qingdao University, Qingdao, Shandong 266071, PR China.
| |
Collapse
|
25
|
Astley RA, Mursalin MH, Coburn PS, Livingston ET, Nightengale JW, Bagaruka E, Hunt JJ, Callegan MC. Ocular Bacterial Infections: A Ten-Year Survey and Review of Causative Organisms Based on the Oklahoma Experience. Microorganisms 2023; 11:1802. [PMID: 37512974 PMCID: PMC10386592 DOI: 10.3390/microorganisms11071802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 06/29/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
Ocular infections can be medical emergencies that result in permanent visual impairment or blindness and loss of quality of life. Bacteria are a major cause of ocular infections. Effective treatment of ocular infections requires knowledge of which bacteria are the likely cause of the infection. This survey of ocular bacterial isolates and review of ocular pathogens is based on a survey of a collection of isolates banked over a ten-year span at the Dean McGee Eye Institute in Oklahoma. These findings illustrate the diversity of bacteria isolated from the eye, ranging from common species to rare and unique species. At all sampled sites, staphylococci were the predominant bacteria isolated. Pseudomonads were the most common Gram-negative bacterial isolate, except in vitreous, where Serratia was the most common Gram-negative bacterial isolate. Here, we discuss the range of ocular infections that these species have been documented to cause and treatment options for these infections. Although a highly diverse spectrum of species has been isolated from the eye, the majority of infections are caused by Gram-positive species, and in most infections, empiric treatments are effective.
Collapse
Affiliation(s)
- Roger A Astley
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Md Huzzatul Mursalin
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Phillip S Coburn
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Erin T Livingston
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - James W Nightengale
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Eddy Bagaruka
- Department of Biology, Oklahoma Christian University, Edmond, OK 73013, USA
| | - Jonathan J Hunt
- Department of Biology, Oklahoma Christian University, Edmond, OK 73013, USA
| | - Michelle C Callegan
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Dean McGee Eye Institute, Oklahoma City, OK 73104, USA
| |
Collapse
|
26
|
Patil R, Dehari D, Chaudhuri A, Kumar DN, Kumar D, Singh S, Nath G, Agrawal AK. Recent advancements in nanotechnology-based bacteriophage delivery strategies against bacterial ocular infections. Microbiol Res 2023; 273:127413. [PMID: 37216845 DOI: 10.1016/j.micres.2023.127413] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 05/16/2023] [Accepted: 05/16/2023] [Indexed: 05/24/2023]
Abstract
Antibiotic resistance is growing as a critical challenge in a variety of disease conditions including ocular infections leading to disastrous effects on the human eyes. Staphylococcus aureus (S. aureus) mediated ocular infections are very common affecting different parts of the eye viz. vitreous chamber, conjunctiva, cornea, anterior and posterior chambers, tear duct, and eyelids. Blepharitis, dacryocystitis, conjunctivitis, keratitis, endophthalmitis, and orbital cellulitis are some of the commonly known ocular infections caused by S. aureus. Some of these infections are so fatal that they could cause bilateral blindness like panophthalmitis and orbital cellulitis, which is caused by methicillin-resistant S. aureus (MRSA) and vancomycin-resistance S. aureus (VRSA). The treatment of S. aureus infections with known antibiotics is becoming gradually difficult because of the development of resistance against multiple antibiotics. Apart from the different combinations and formulation strategies, bacteriophage therapy is growing as an effective alternative to treat such infections. Although the superiority of bacteriophage therapy is well established, yet physical factors (high temperatures, acidic pH, UV-rays, and ionic strength) and pharmaceutical barriers (poor stability, low in-vivo retention, controlled and targeted delivery, immune system neutralization, etc.) have the greatest influence on the viability of phage virions (also phage proteins). A variety of Nanotechnology based formulations such as polymeric nanoparticles, liposomes, dendrimers, nanoemulsions, and nanofibres have been recently reported to overcome the above-mentioned obstacles. In this review, we have compiled all these recent reports and discussed bacteriophage-based nanoformulations techniques for the successful treatment of ocular infections caused by multidrug-resistant S. aureus and other bacteria.
Collapse
Affiliation(s)
- Rohit Patil
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, U.P., India
| | - Deepa Dehari
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, U.P., India
| | - Aiswarya Chaudhuri
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, U.P., India
| | - Dulla Naveen Kumar
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, U.P., India
| | - Dinesh Kumar
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, U.P., India
| | - Sanjay Singh
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, U.P., India; Babasaheb Bhimrao Ambedkar University, Lucknow 226025, U.P., India
| | - Gopal Nath
- Department of Microbiology, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, U.P., India
| | - Ashish Kumar Agrawal
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, U.P., India.
| |
Collapse
|
27
|
Englisch CN, Wadood NA, Pätzold L, Gallagher A, Krasteva-Christ G, Becker SL, Bischoff M. Establishing an Experimental Pseudomonas aeruginosa Keratitis Model in Mice - Challenges and Solutions. Ann Anat 2023; 249:152099. [PMID: 37105406 DOI: 10.1016/j.aanat.2023.152099] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 03/27/2023] [Accepted: 03/28/2023] [Indexed: 04/29/2023]
Abstract
BACKGROUND With the ongoing increase in antimicrobial resistances seen in bacterial isolates causing a keratitis in humans, animal models have become an important tool to study new antimicrobial therapies. Nevertheless, the establishment of experimental keratitis is difficult. Here, we discuss the impact of different arrangements, including animal age, bacterial strain and dose as well as epithelium removal on the outcome of experimental keratitis. We therefore present the methods and results of our establishing experiments. METHODS Bacterial load determination and flow cytometry were performed using eye homogenate gained from a 72hours lasting murine Pseudomonas aeruginosa keratitis model. Additionally, the intensity of the infection was scored from 0 to 5, the mice weighed, and blood immune cells counted. RESULTS We found that older C57BL/6N mice (8-11 months) are more susceptible to develop a keratitis than younger mice (5-6 weeks). Epithelium removal has no major impact on infectivity and disease progression in aged mice. P. aeruginosa exoU+ strains, such as PA54, should preferentially be used and highly concentrated (∼ 5×107 CFU). Establishing an infection with the exoU- PAO1 derivative DSM 19880 was not possible. CONCLUSIONS We present a replicable method to achieve a successful experimental P. aeruginosa keratitis in C57BL/6N mice that is sustained or aggravated over the observation period of 3 days in 80% of all animals tested. Our work is of particular interest to all researchers planning the establishment of such experimental models. We show some key aspects that can simplify and quicken the procedure, ultimately saving costs and animal life.
Collapse
Affiliation(s)
- Colya N Englisch
- Institute for Medical Microbiology and Hygienics, Saarland University, 66421, Homburg/Saar, Germany.
| | - Noran Abdel Wadood
- Institute for Medical Microbiology and Hygienics, Saarland University, 66421, Homburg/Saar, Germany; Institute of Anatomy and Cell Biology, Saarland University, 66421, Homburg/Saar, Germany.
| | - Linda Pätzold
- Institute for Medical Microbiology and Hygienics, Saarland University, 66421, Homburg/Saar, Germany.
| | | | | | - Sören L Becker
- Institute for Medical Microbiology and Hygienics, Saarland University, 66421, Homburg/Saar, Germany.
| | - Markus Bischoff
- Institute for Medical Microbiology and Hygienics, Saarland University, 66421, Homburg/Saar, Germany.
| |
Collapse
|
28
|
Ung L, Chodosh J. Urgent unmet needs in the care of bacterial keratitis: An evidence-based synthesis. Ocul Surf 2023; 28:378-400. [PMID: 34461290 PMCID: PMC10721114 DOI: 10.1016/j.jtos.2021.08.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 08/18/2021] [Accepted: 08/19/2021] [Indexed: 11/21/2022]
Abstract
Bacterial corneal infections, or bacterial keratitis (BK), are ophthalmic emergencies that frequently lead to irreversible visual impairment. Though increasingly recognized as a major cause of global blindness, modern paradigms of evidence-based care in BK have remained at a diagnostic and therapeutic impasse for over half a century. Current standards of management - based on the collection of corneal cultures and the application of broad-spectrum topical antibiotics - are beset by important yet widely underrecognized limitations, including approximately 30% of all patients who will develop moderate to severe vision loss in the affected eye. Though recent advances have involved a more clearly defined role for adjunctive topical corticosteroids, and novel therapies such as corneal crosslinking, overall progress to improve patient and population-based outcomes remains incommensurate to the chronic morbidity caused by this disease. Recognizing that the care of BK is guided by the clinical axiom, "time equals vision", this chapter offers an evidence-based synthesis for the clinical management of these infections, underscoring critical unmet needs in disease prevention, diagnosis, and treatment.
Collapse
Affiliation(s)
- Lawson Ung
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA; Infectious Disease Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA; Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - James Chodosh
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA; Infectious Disease Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
29
|
Ramadan A, Cao Z, Hassan M, Zetterberg F, Nilsson UJ, Gadjeva M, Rathinam V, Panjwani N. Galectin-8 Downmodulates TLR4 Activation and Impairs Bacterial Clearance in a Mouse Model of Pseudomonas aeruginosa Keratitis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:398-407. [PMID: 36603009 PMCID: PMC9898164 DOI: 10.4049/jimmunol.2200706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 11/15/2022] [Indexed: 01/06/2023]
Abstract
Pseudomonas aeruginosa provokes a painful, sight-threatening corneal infection. It progresses rapidly and is difficult to treat. In this study, using a mouse model of P. aeruginosa keratitis, we demonstrate the importance of a carbohydrate-binding protein, galectin-8 (Gal-8), in regulation of the innate immune response. First, using two distinct strains of P. aeruginosa, we established that Gal-8-/- mice are resistant to P. aeruginosa keratitis. In contrast, mice deficient in Gal-1, Gal-3, and Gal-9 were fully susceptible. Second, the addition of exogenous rGal-8 to LPS (TLR4 ligand)-stimulated bone marrow-derived macrophages suppressed 1) the activation of the NF-κB pathway, and 2) formation of the MD-2/TLR4 complex. Additionally, the expression level of reactive oxygen species was substantially higher in infected Gal-8-/- bone marrow neutrophils (BMNs) compared with the Gal-8+/+ BMNs, and the P. aeruginosa killing capacity of Gal-8-/- BMNs was considerably higher compared with that of Gal-8+/+ BMNs. In the bacterial killing assays, the addition of exogenous rGal-8 almost completely rescued the phenotype of Gal-8-/- BMNs. Finally, we demonstrate that a subconjunctival injection of a Gal-8 inhibitor markedly reduces the severity of infection in the mouse model of P. aeruginosa keratitis. These data lead us to conclude that Gal-8 downmodulates the innate immune response by suppressing activation of the TLR4 pathway and clearance of P. aeruginosa by neutrophils. These findings have broad implications for developing novel therapeutic strategies for treatment of conditions resulting from the hyperactive immune response both in ocular as well as nonocular tissues.
Collapse
Affiliation(s)
- Abdulraouf Ramadan
- New England Eye Center/Department of Ophthalmology, Tufts University School of Medicine, Boston, MA. 02111, USA
| | - Zhiyi Cao
- New England Eye Center/Department of Ophthalmology, Tufts University School of Medicine, Boston, MA. 02111, USA
| | - Mujtaba Hassan
- Centre for Analysis and Synthesis, Department of Chemistry, Lund University, Lund, Sweden
| | | | - Ulf J. Nilsson
- Centre for Analysis and Synthesis, Department of Chemistry, Lund University, Lund, Sweden
| | - Mihaela Gadjeva
- Department of Medicine, Division of Infectious Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA. 02115, USA
| | - Vijay Rathinam
- Department of Immunology, UConn Health School of Medicine, Farmington, CT
| | - Noorjahan Panjwani
- New England Eye Center/Department of Ophthalmology, Tufts University School of Medicine, Boston, MA. 02111, USA
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA, 02111, USA
| |
Collapse
|
30
|
Silva MD, André C, Bispo PJ. Targeted Killing of Ocular Streptococcus pneumoniae by the Phage Endolysin MSlys. OPHTHALMOLOGY SCIENCE 2022; 2:100193. [PMID: 36531571 PMCID: PMC9754957 DOI: 10.1016/j.xops.2022.100193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Affiliation(s)
- Maria Daniela Silva
- CEB−Centre of Biological Engineering, University of Minho, Braga, Portugal
- Department of Otolaryngology, Massachusetts Eye and Ear, Boston, Massachusetts
| | - Camille André
- Department of Ophthalmology, Massachusetts Eye and Ear, and Infectious Disease Institute, Harvard Medical School, Boston, Massachusetts
| | - Paulo J.M. Bispo
- Department of Ophthalmology, Massachusetts Eye and Ear, and Infectious Disease Institute, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
31
|
Deep Learning Approach in Image Diagnosis of Pseudomonas Keratitis. Diagnostics (Basel) 2022; 12:diagnostics12122948. [PMID: 36552954 PMCID: PMC9777188 DOI: 10.3390/diagnostics12122948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 11/15/2022] [Accepted: 11/22/2022] [Indexed: 11/27/2022] Open
Abstract
This investigation aimed to explore deep learning (DL) models' potential for diagnosing Pseudomonas keratitis using external eye images. In the retrospective research, the images of bacterial keratitis (BK, n = 929), classified as Pseudomonas (n = 618) and non-Pseudomonas (n = 311) keratitis, were collected. Eight DL algorithms, including ResNet50, DenseNet121, ResNeXt50, SE-ResNet50, and EfficientNets B0 to B3, were adopted as backbone models to train and obtain the best ensemble 2-, 3-, 4-, and 5-DL models. Five-fold cross-validation was used to determine the ability of single and ensemble models to diagnose Pseudomonas keratitis. The EfficientNet B2 model had the highest accuracy (71.2%) of the eight single-DL models, while the best ensemble 4-DL model showed the highest accuracy (72.1%) among the ensemble models. However, no statistical difference was shown in the area under the receiver operating characteristic curve and diagnostic accuracy among these single-DL models and among the four best ensemble models. As a proof of concept, the DL approach, via external eye photos, could assist in identifying Pseudomonas keratitis from BK patients. All the best ensemble models can enhance the performance of constituent DL models in diagnosing Pseudomonas keratitis, but the enhancement effect appears to be limited.
Collapse
|
32
|
Antibacterial Activity of Antibiotic-Releasing Polydopamine-Coated Nephrite Composites for Application in Drug-Eluting Contact Lens. MATERIALS 2022; 15:ma15144823. [PMID: 35888290 PMCID: PMC9315558 DOI: 10.3390/ma15144823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 07/05/2022] [Accepted: 07/07/2022] [Indexed: 11/17/2022]
Abstract
The aim of this study is to prepare ciprofloxacin (CIP) or levofloxacin (LEVO)-incorporated and polydopamine (PDA)-coated nephrite composites for application in drug-eluting contact lenses. PDA was coated onto the surface of nephrite to improve antibacterial activity and to payload antibiotics. CIP or LEVO was incorporated into the PDA layer on the surface of nephrite. Furthermore, CIP-incorporated/PDA-coated nephrite composites were embedded into the contact lenses. PDA-coated nephrite composites showed dull and smooth surfaces according to the dopamine concentration while nephrite itself has sharp surface morphology. CIP- or LEVO-loaded/PDA-coated nephrite composites also have dull and smooth surface properties. Nano and/or sub-micron clusters were observed in field emission-scanning electron microscopy (FE-SEM) observation, indicating that PDA nanoparticles were accumulated and coated onto the surface of nephrite. Furthermore, CIP- or LEVO-incorporated/PDA-coated nephrite composites showed the sustained release of CIP or LEVO in vitro and these properties contributed to the enhanced antibacterial activity of composites compared to nephrite or PDA-coated nephrite composites. CIP-incorporated/PDA-coated nephrite composites were embedded in the contact lenses and then, in an antibacterial study, they showed higher bactericidal effect against Staphylococcus aureus (S. aureus) compared to nephrite itself or PDA-coated nephrite composites. We suggest that CIP- or LEVO-loaded/PDA-coated nephrite composite-embedded contact lenses are a promising candidate for therapeutic application.
Collapse
|
33
|
Cabrera‐Aguas M, Khoo P, Watson SL. Infectious keratitis: A review. Clin Exp Ophthalmol 2022; 50:543-562. [PMID: 35610943 PMCID: PMC9542356 DOI: 10.1111/ceo.14113] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 05/05/2022] [Accepted: 05/06/2022] [Indexed: 12/29/2022]
Abstract
Globally, infectious keratitis is the fifth leading cause of blindness. The main predisposing factors include contact lens wear, ocular injury and ocular surface disease. Staphylococcus species, Pseudomonas aeruginosa, Fusarium species, Candida species and Acanthamoeba species are the most common causal organisms. Culture of corneal scrapes is the preferred initial test to identify the culprit organism. Polymerase chain reaction (PCR) tests and in vivo confocal microscopy can complement the diagnosis. Empiric therapy is typically commenced with fluoroquinolones, or fortified antibiotics for bacterial keratitis; topical natamycin for fungal keratitis; and polyhexamethylene biguanide or chlorhexidine for acanthamoeba keratitis. Herpes simplex keratitis is mainly diagnosed clinically; however, PCR can also be used to confirm the initial diagnosis and in atypical cases. Antivirals and topical corticosteroids are indicated depending on the corneal layer infected. Vision impairment, blindness and even loss of the eye can occur with a delay in diagnosis and inappropriate antimicrobial therapy.
Collapse
Affiliation(s)
- Maria Cabrera‐Aguas
- Save Sight Institute, Discipline of Ophthalmology, Faculty of Medicine and Health The University of Sydney Sydney New South Wales Australia
- Corneal Unit Sydney Eye Hospital Sydney New South Wales Australia
| | - Pauline Khoo
- Save Sight Institute, Discipline of Ophthalmology, Faculty of Medicine and Health The University of Sydney Sydney New South Wales Australia
- Corneal Unit Sydney Eye Hospital Sydney New South Wales Australia
| | - Stephanie L. Watson
- Save Sight Institute, Discipline of Ophthalmology, Faculty of Medicine and Health The University of Sydney Sydney New South Wales Australia
- Corneal Unit Sydney Eye Hospital Sydney New South Wales Australia
| |
Collapse
|
34
|
Ting DSJ, Mohammed I, Lakshminarayanan R, Beuerman RW, Dua HS. Host Defense Peptides at the Ocular Surface: Roles in Health and Major Diseases, and Therapeutic Potentials. Front Med (Lausanne) 2022; 9:835843. [PMID: 35783647 PMCID: PMC9243558 DOI: 10.3389/fmed.2022.835843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 05/17/2022] [Indexed: 11/13/2022] Open
Abstract
Sight is arguably the most important sense in human. Being constantly exposed to the environmental stress, irritants and pathogens, the ocular surface – a specialized functional and anatomical unit composed of tear film, conjunctival and corneal epithelium, lacrimal glands, meibomian glands, and nasolacrimal drainage apparatus – serves as a crucial front-line defense of the eye. Host defense peptides (HDPs), also known as antimicrobial peptides, are evolutionarily conserved molecular components of innate immunity that are found in all classes of life. Since the first discovery of lysozyme in 1922, a wide range of HDPs have been identified at the ocular surface. In addition to their antimicrobial activity, HDPs are increasingly recognized for their wide array of biological functions, including anti-biofilm, immunomodulation, wound healing, and anti-cancer properties. In this review, we provide an updated review on: (1) spectrum and expression of HDPs at the ocular surface; (2) participation of HDPs in ocular surface diseases/conditions such as infectious keratitis, conjunctivitis, dry eye disease, keratoconus, allergic eye disease, rosacea keratitis, and post-ocular surgery; (3) HDPs that are currently in the development pipeline for treatment of ocular diseases and infections; and (4) future potential of HDP-based clinical pharmacotherapy for ocular diseases.
Collapse
Affiliation(s)
- Darren Shu Jeng Ting
- Academic Ophthalmology, School of Medicine, University of Nottingham, Nottingham, United Kingdom
- Department of Ophthalmology, Queen's Medical Centre, Nottingham, United Kingdom
- Anti-Infectives Research Group, Singapore Eye Research Institute, Singapore, Singapore
- *Correspondence: Darren Shu Jeng Ting
| | - Imran Mohammed
- Academic Ophthalmology, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | | | - Roger W. Beuerman
- Anti-Infectives Research Group, Singapore Eye Research Institute, Singapore, Singapore
| | - Harminder S. Dua
- Academic Ophthalmology, School of Medicine, University of Nottingham, Nottingham, United Kingdom
- Department of Ophthalmology, Queen's Medical Centre, Nottingham, United Kingdom
| |
Collapse
|
35
|
Ung L, Chodosh J. COVID-19 and the eye: alternative facts The 2022 Bowman Club, David L. Easty lecture. BMJ Open Ophthalmol 2022; 7:bmjophth-2022-001042. [PMID: 35675203 PMCID: PMC9114314 DOI: 10.1136/bmjophth-2022-001042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 04/18/2022] [Indexed: 01/08/2023] Open
Abstract
In addition to catastrophic loss of life, and dramatic and unwanted alterations to the daily lives of those left behind, the COVID-19 pandemic has fostered the publication and dissemination of an unprecedented quantity of peer-reviewed medical and scientific publications on a single subject. In particular, the ophthalmic literature is now replete with clinical and laboratory studies on putative eye involvement by SARS-CoV-2, the aetiologic agent of COVID-19. In this review, we critically appraise the published literature on COVID-19, and suggest that the quality of scientific peer review and editorial decision-making also suffered during the COVID-19 pandemic.
Collapse
Affiliation(s)
- Lawson Ung
- Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, USA
- Department of Epidemiology, Harvard University T. H. Chan School of Public Health, Boston, Massachusetts, USA
| | - James Chodosh
- Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
36
|
Singh RB, Das S, Chodosh J, Sharma N, Zegans ME, Kowalski RP, Jhanji V. Paradox of complex diversity: Challenges in the diagnosis and management of bacterial keratitis. Prog Retin Eye Res 2021; 88:101028. [PMID: 34813978 DOI: 10.1016/j.preteyeres.2021.101028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 11/09/2021] [Accepted: 11/12/2021] [Indexed: 12/12/2022]
Abstract
Bacterial keratitis continues to be one of the leading causes of corneal blindness in the developed as well as the developing world, despite swift progress since the dawn of the "anti-biotic era". Although, we are expeditiously developing our understanding about the different causative organisms and associated pathology leading to keratitis, extensive gaps in knowledge continue to dampen the efforts for early and accurate diagnosis, and management in these patients, resulting in poor clinical outcomes. The ability of the causative bacteria to subdue the therapeutic challenge stems from their large genome encoding complex regulatory networks, variety of unique virulence factors, and rapid secretion of tissue damaging proteases and toxins. In this review article, we have provided an overview of the established classical diagnostic techniques and therapeutics for keratitis caused by various bacteria. We have extensively reported our recent in-roads through novel tools for accurate diagnosis of mono- and poly-bacterial corneal infections. Furthermore, we outlined the recent progress by our group and others in understanding the sub-cellular genomic changes that lead to antibiotic resistance in these organisms. Finally, we discussed in detail, the novel therapies and drug delivery systems in development for the efficacious management of bacterial keratitis.
Collapse
Affiliation(s)
- Rohan Bir Singh
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA; Department of Ophthalmology, Leiden University Medical Center, 2333, ZA Leiden, the Netherlands
| | - Sujata Das
- Cornea and Anterior Segment Services, LV Prasad Eye Institute, Bhubaneshwar, India
| | - James Chodosh
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Namrata Sharma
- Dr. Rajendra Prasad Centre for Ophthalmic Sciences, All India Institute of Medical Sciences, New Delhi, India
| | - Michael E Zegans
- Department of Ophthalmology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Regis P Kowalski
- Department of Ophthalmology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA; The Charles T Campbell Ophthalmic Microbiology Laboratory, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Vishal Jhanji
- Department of Ophthalmology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA; The Charles T Campbell Ophthalmic Microbiology Laboratory, University of Pittsburgh Medical Center, Pittsburgh, PA, USA.
| |
Collapse
|
37
|
Nishida T, Sugioka K, Fukuda K, Murakami J. Pivotal Role of Corneal Fibroblasts in Progression to Corneal Ulcer in Bacterial Keratitis. Int J Mol Sci 2021; 22:ijms22168979. [PMID: 34445684 PMCID: PMC8396668 DOI: 10.3390/ijms22168979] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 08/12/2021] [Accepted: 08/17/2021] [Indexed: 01/05/2023] Open
Abstract
The shape and transparency of the cornea are essential for clear vision. However, its location at the ocular surface renders the cornea vulnerable to pathogenic microorganisms in the external environment. Pseudomonas aeruginosa and Staphylococcus aureus are two such microorganisms and are responsible for most cases of bacterial keratitis. The development of antimicrobial agents has allowed the successful treatment of bacterial keratitis if the infection is diagnosed promptly. However, no effective medical treatment is available after progression to corneal ulcer, which is characterized by excessive degradation of collagen in the corneal stroma and can lead to corneal perforation and corneal blindness. This collagen degradation is mediated by both infecting bacteria and corneal fibroblasts themselves, with a urokinase-type plasminogen activator (uPA)-plasmin-matrix metalloproteinase (MMP) cascade playing a central role in collagen destruction by the host cells. Bacterial factors stimulate the production by corneal fibroblasts of both uPA and pro-MMPs, released uPA mediates the conversion of plasminogen in the extracellular environment to plasmin, and plasmin mediates the conversion of secreted pro-MMPs to the active form of these enzymes, which then degrade stromal collagen. Bacterial factors also stimulate expression by corneal fibroblasts of the chemokine interleukin-8 and the adhesion molecule ICAM-1, both of which contribute to recruitment and activation of polymorphonuclear neutrophils, and these cells then further stimulate corneal fibroblasts via the secretion of interleukin-1. At this stage of the disease, bacteria are no longer necessary for collagen degradation. In this review, we discuss the pivotal role of corneal fibroblasts in corneal ulcer associated with infection by P. aeruginosa or S. aureus as well as the development of potential new modes of treatment for this condition.
Collapse
Affiliation(s)
- Teruo Nishida
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755-8505, Japan;
- Division of Cornea and Ocular Surface, Ohshima Eye Hospital, Fukuoka 812-0036, Japan
| | - Koji Sugioka
- Department of Ophthalmology, Kindai University Nara Hospital, Ikoma, Nara 630-0293, Japan;
| | - Ken Fukuda
- Department of Ophthalmology and Visual Science, Kochi Medical School, Kochi University, Nankoku, Kochi 783-8505, Japan
- Correspondence:
| | - Junko Murakami
- Division of Ophthalmology, Sakibana Hospital, Izumi, Osaka 594-1105, Japan;
| |
Collapse
|