1
|
Dong L, Fan Z, Fang B, Zhao X, Yao H, Cai G, Yang S, Zhang G, Cheng X, Feng Y, Mi S, Sun W. Oriented cellulose hydrogel: Directed tissue regeneration for reducing corneal leukoplakia and managing fungal corneal ulcers. Bioact Mater 2024; 41:15-29. [PMID: 39101028 PMCID: PMC11292264 DOI: 10.1016/j.bioactmat.2024.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 06/18/2024] [Accepted: 07/04/2024] [Indexed: 08/06/2024] Open
Abstract
Fungal corneal ulcer is one of the leading causes of corneal blindness in developing countries. Corneal scars such as leukoplakia are formed due to inflammation, oxidative stress and non-directed repair, which seriously affect the patients' subsequent visual and life quality. In this study, drawing inspiration from the oriented structure of collagen fibers within the corneal stroma, we first proposed the directional arrangement of CuTA-CMHT hydrogel system at micro and macro scales based on the 3D printing extrusion method combined with secondary patterning. It played an antifungal role and induced oriented repair in therapy of fungal corneal ulcer. The results showed that it effectively inhibited Candida albicans, Aspergillus Niger, Fusarium sapropelum, which mainly affects TNF, NF-kappa B, and HIF-1 signaling pathways, achieving effective antifungal functions. More importantly, the fibroblasts interacted with extracellular matrix (ECM) of corneal stroma through formation of focal adhesions, promoted the proliferation and directional migration of cells in vitro, induced the directional alignment of collagen fibers and corneal stromal orthogonally oriented repair in vivo. This process is mainly associated with MYLK, MYL9, and ITGA3 molecules. Furthermore, the downregulation the growth factors TGF-β and PDGF-β inhibits myofibroblast development and reduces scar-type ECM production, thereby reducing corneal leukoplakia. It also activates the PI3K-AKT signaling pathway, promoting corneal healing. In conclusion, the oriented CuTA-CMHT hydrogel system mimics the orthogonal arrangement of collagen fibers, inhibits inflammation, eliminates reactive oxygen species, and reduces corneal leukoplakia, which is of great significance in the treatment of fungal corneal ulcer and is expected to write a new chapter in corneal tissue engineering.
Collapse
Affiliation(s)
- Lina Dong
- Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China
- Department of Burns, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, China
| | - Zixin Fan
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, Shenzhen, Guangdong, 518040, China
| | - Bixing Fang
- Department of Otolaryngology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, China
| | - Xiaoyu Zhao
- Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China
| | - Hongyi Yao
- Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China
| | - Gangpei Cai
- Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China
| | - Shuo Yang
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, Shenzhen, Guangdong, 518040, China
| | - Guoming Zhang
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, Shenzhen, Guangdong, 518040, China
| | - Xiaoqi Cheng
- Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China
| | - Yun Feng
- Department of Ophthalmology, Peking University Third Hospital, Beijing, 100191, China
| | - Shengli Mi
- Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China
| | - Wei Sun
- Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China
- Department of Mechanical Engineering, Biomanufacturing Center, Tsinghua University, Beijing, China
- Department of Mechanical Engineering, Drexel University, Philadelphia, PA, USA
| |
Collapse
|
2
|
Fu X, Tian X, Lin J, Wang Q, Gu L, Wang Z, Chi M, Yu B, Feng Z, Liu W, Zhang L, Li C, Zhao G. Zeolitic Imidazolate Framework-8 Offers an Anti-Inflammatory and Antifungal Method in the Treatment of Aspergillus Fungal Keratitis in vitro and in vivo. Int J Nanomedicine 2024; 19:11163-11179. [PMID: 39502641 PMCID: PMC11537184 DOI: 10.2147/ijn.s480800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 10/26/2024] [Indexed: 11/08/2024] Open
Abstract
Background Fungal keratitis is a serious blinding eye disease. Traditional drugs used to treat fungal keratitis commonly have the disadvantages of low bioavailability, poor dispersion, and limited permeability. Purpose To develop a new method for the treatment of fungal keratitis with improved bioavailability, dispersion, and permeability. Methods Zeolitic Imidazolate Framework-8 (ZIF-8) was formed by zinc ions and 2-methylimidazole linked by coordination bonds and characterized by Scanning electron microscopy (SEM), X-ray diffraction (XRD), and Zeta potential. The safety of ZIF-8 on HCECs and RAW 264.7 cells was detected by Cell Counting Kit-8 (CCK-8). Safety evaluation of ZIF-8 on mice corneal epithelium was conducted using the Draize corneal toxicity test. The effects of ZIF-8 on fungal growth, biofilm formation, and hyphae structure were detected by Minimal inhibit concentration (MIC), crystal violet staining, Propidium Iodide (PI) testing, and calcofluor white staining. The anti-inflammatory effects of ZIF-8 on RAW 246.7 cells were evaluated by Quantitative Real-Time PCR Experiments (qPCR) and Enzyme-linked immunosorbent assay (ELISA). Clinical score, Colony-Forming Units (CFU), Hematoxylin-eosin (HE) staining, and immunofluorescence were conducted to verify the therapeutic effect of ZIF-8 on C57BL/6 female mice with fungal keratitis. Results In vitro, ZIF-8 showed outstanding antifungal effects, including inhibiting the growth of Aspergillus fumigatus over 90% at 64 μg/mL, restraining the formation of biofilm, and destroying cell membranes. In vivo, treatment with ZIF-8 reduced corneal fungal load and mitigated neutrophil infiltration in fungal keratitis, which effectively reduced the severity of keratitis in mice and alleviated the infiltration of inflammatory factors in the mouse cornea. In addition, ZIF-8 reduces the inflammatory response by downregulating the expression of pro-inflammatory cytokines TNF-α, IL-6, and IL-1β after Aspergillus fumigatus infection in vivo and in vitro. Conclusion ZIF-8 has a significant anti-inflammatory and antifungal effect, which provides a new solution for the treatment of fungal keratitis.
Collapse
Affiliation(s)
- Xueyun Fu
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Xue Tian
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Jing Lin
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Qian Wang
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Lingwen Gu
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Ziyi Wang
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Menghui Chi
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Bing Yu
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Zhuhui Feng
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Wenyao Liu
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Lina Zhang
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Cui Li
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Guiqiu Zhao
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| |
Collapse
|
3
|
Datta D, Priyanka Bandi S, Colaco V, Dhas N, Siva Reddy DV, Vora LK. Fostering the unleashing potential of nanocarriers-mediated delivery of ocular therapeutics. Int J Pharm 2024; 658:124192. [PMID: 38703931 DOI: 10.1016/j.ijpharm.2024.124192] [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: 03/08/2024] [Revised: 04/21/2024] [Accepted: 04/30/2024] [Indexed: 05/06/2024]
Abstract
Ocular delivery is the most challenging aspect in the field of pharmaceutical research. The major hurdle for the controlled delivery of drugs to the eye includes the physiological static barriers such as the complex layers of the cornea, sclera and retina which restrict the drug from permeating into the anterior and posterior segments of the eye. Recent years have witnessed inventions in the field of conventional and nanocarrier drug delivery which have shown considerable enhancement in delivering small to large molecules across the eye. The dynamic challenges associated with conventional systems include limited drug contact time and inadequate ocular bioavailability resulting from solution drainage, tear turnover, and dilution or lacrimation. To this end, various bioactive-based nanosized carriers including liposomes, ethosomes, niosomes, dendrimer, nanogel, nanofibers, contact lenses, nanoprobes, selenium nanobells, nanosponge, polymeric micelles, silver nanoparticles, and gold nanoparticles among others have been developed to circumvent the limitations associated with the conventional dosage forms. These nanocarriers have been shown to achieve enhanced drug permeation or retention and prolong drug release in the ocular tissue due to their better tissue adherence. The surface charge and the size of nanocarriers (10-1000 nm) are the important key factors to overcome ocular barriers. Various nanocarriers have been shown to deliver active therapeutic molecules including timolol maleate, ampicillin, natamycin, voriconazole, cyclosporine A, dexamethasone, moxifloxacin, and fluconazole among others for the treatment of anterior and posterior eye diseases. Taken together, in a nutshell, this extensive review provides a comprehensive perspective on the numerous facets of ocular drug delivery with a special focus on bioactive nanocarrier-based approaches, including the difficulties and constraints involved in the fabrication of nanocarriers. This also provides the detailed invention, applications, biodistribution and safety-toxicity of nanocarriers-based therapeutcis for the ophthalmic delivery.
Collapse
Affiliation(s)
- Deepanjan Datta
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India.
| | - Sony Priyanka Bandi
- Loka Laboratories Private Limited, Technology Business Incubator, BITS Pilani Hyderabad Campus, Jawahar Nagar, Medchal 500078, Telangana, India.
| | - Viola Colaco
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | - Namdev Dhas
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | - D V Siva Reddy
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio TX78227, USA
| | - Lalitkumar K Vora
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, U.K
| |
Collapse
|
4
|
Zhan L, Tian X, Lin J, Zhang Y, Zhao G, Peng X. The Therapeutic Role and Mechanism of Glabridin Under Aspergillus fumigatus Infection. J Ocul Pharmacol Ther 2024; 40:89-99. [PMID: 38346287 DOI: 10.1089/jop.2023.0085] [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] [Indexed: 02/15/2024] Open
Abstract
Purpose: To characterize the efficiency of glabridin alone and in combination with clinical antifungals in Aspergillus fumigatus keratitis. Methods: The broth microdilution method was performed to investigate whether glabridin exerted an antifungal role on planktonic cells and immature and mature biofilm. Antifungal mechanism was evaluated by Sorbitol and Ergosterol Assays. The synergistic effect of glabridin and antifungals was assessed through the checkerboard microdilution method and time-killing test. Regarding anti-inflammatory role, inflammatory substances induced by A. fumigatus were assessed by real-time quantitative polymerase chain reaction, western blot, and enzyme-linked immunosorbent assay. Drug toxicity was assessed by Draize test in vivo. Macrophage phenotypes were examined by flow cytometry. Results: Regarding antifungal activity, glabridin destroyed fungal cell wall and membrane on planktonic cells and suppressed immature and mature biofilm formation. After combining with natamycin or amphotericin B, glabridin possessed a potent synergistic effect against A. fumigatus. Regarding anti-inflammatory aspects, Dectin-1, toll‑like receptor (TLR)-2 and TLR-4 expression of human corneal epithelial cells were significantly elevated after A. fumigatus challenge and reduced by glabridin. The elevated expression of interleukin-1β and tumor necrosis factor-alpha induced by A. fumigatus or corresponding agonists were reversed by glabridin, equivalent to the effect of corresponding inhibitors. Glabridin could also contribute to anti-inflammation by downregulating inflammatory mediator expression to suppress macrophage infiltration. Conclusions: Glabridin contributed to fungal clearance by destroying fungal cell wall and membrane, and disrupting biofilm. Combining glabridin with clinical antifungals was superior in reducing A. fumigatus growth. Glabridin exerted an anti-inflammatory effect by downregulating proinflammatory substance expression and inhibiting macrophage infiltration, which provide a potential agent and treatment strategies for fungal keratitis.
Collapse
Affiliation(s)
- Lu Zhan
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xue Tian
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jing Lin
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yingxue Zhang
- Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Guiqiu Zhao
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xudong Peng
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
- Department of Ophthalmology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
5
|
Madkhali OA. Drug Delivery of Gelatin Nanoparticles as a Biodegradable Polymer for the Treatment of Infectious Diseases: Perspectives and Challenges. Polymers (Basel) 2023; 15:4327. [PMID: 37960007 PMCID: PMC10648051 DOI: 10.3390/polym15214327] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 10/31/2023] [Accepted: 11/03/2023] [Indexed: 11/15/2023] Open
Abstract
In recent years, there has been a growing interest in the use of gelatin nanoparticles (GNPs) for the treatment of infectious diseases. The inherent properties of these nanoparticles make them attractive options for drug delivery. Their biocompatibility ensures that they can interact with biological systems without causing adverse reactions, while their biodegradability ensures that they can break down harmlessly in the body once their function is performed. Furthermore, their capacity for controlled drug release ensures that therapeutic agents can be delivered over a sustained period, thereby enhancing treatment efficacy. This review examines the current landscape of GNP-based drug delivery, with a specific focus on its potential applications and challenges in the context of infectious diseases. Key challenges include controlling drug release rates, ensuring nanoparticle stability under physiological conditions, scaling up production while maintaining quality, mitigating potential immunogenic reactions, optimizing drug loading efficiency, and tracking the biodistribution and clearance of GNPs in the body. Despite these hurdles, GNPs hold promising potential in the realm of infectious disease treatment. Ongoing research and innovation are essential to overcome these obstacles and completely harness the potential of GNPs in clinical applications.
Collapse
Affiliation(s)
- Osama A Madkhali
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Jazan 45124, Saudi Arabia
| |
Collapse
|
6
|
Wang H, Yin X, Zhang Z, Wang Y, Zhang L, Guo J, Li M. Evaluation of 0.01% Hypochlorous Acid Eye Drops Combined with Conventional Treatment in the Management of Fungal Corneal Ulcers: Randomized Controlled Trial. Curr Eye Res 2023; 48:887-893. [PMID: 37493085 DOI: 10.1080/02713683.2023.2226374] [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: 06/30/2022] [Accepted: 06/13/2023] [Indexed: 07/27/2023]
Abstract
PURPOSE To evaluate the efficacy and safety of hypochlorous acid (HOCI) eye drops in the treatment of fungal keratitis. METHODS A total of 96 patients (96 eyes) with fungal keratitis were randomly divided into two groups: group Ι (conventional treatment + topical HOCI eye drops); The group II (conventional treatment). According to its severity, those patients were divided into grade Ι or grade II. Use of fungal scraping and culture to identify the type of fungal infection, slit lamp examination, and corneal fluorescein staining to observe regression, and confocal corneal microscopy to assess fungal mycelial changes. The main outcome measures were the success rate, healing time, visual recovery, and complications. The Kaplan-Meier curve method was used to analysis of the survival function of days to cure between the two groups. RESULTS There were no statistical differences between the two groups in terms of general condition, medical history, and grading. Corneal scraping results showed that all patients had filamentous fungi. For grade Ι patients, all patients were cured, and the patients in Group I showed faster healing speed than that in Group II (t = -3.665, p < .01). For grade II patients, the recovery time (t = -4.121, p < .01) and the disappearance of hypopyon (t = -4.291, p < .01) were significantly faster in the combination group. In grade Ι and II patients, the final visual acuity and the incidence of complications such as corneal neovascularization, cataract, and hyphema showed no differences in both groups. The survival curve showed that the healing rate of ulcers in the combination treatment group was faster than that in the conventional treatment group (χ2 = 14.332, p = .001). CONCLUSION HOCI can accelerate the healing of fungal keratitis without obvious complications, indicating a promising future in the field of keratitis treatment.
Collapse
Affiliation(s)
- He Wang
- Department of Ophthalmology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Xiaoyue Yin
- Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Zhaowei Zhang
- Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Yining Wang
- Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Ling Zhang
- Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Jianxin Guo
- Department of Ophthalmology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Mingxin Li
- Department of Ophthalmology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| |
Collapse
|
7
|
Jin M, He B, Cai X, Lei Z, Sun T. Research progress of nanoparticle targeting delivery systems in bacterial infections. Colloids Surf B Biointerfaces 2023; 229:113444. [PMID: 37453264 DOI: 10.1016/j.colsurfb.2023.113444] [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: 04/12/2023] [Revised: 06/28/2023] [Accepted: 07/05/2023] [Indexed: 07/18/2023]
Abstract
Bacterial infection is a huge threat to the health of human beings and animals. The abuse of antibiotics have led to the occurrence of bacterial multidrug resistance, which have become a difficult problem in the treatment of clinical infections. Given the outstanding advantages of nanodrug delivery systems in cancer treatment, many scholars have begun to pay attention to their application in bacterial infections. However, due to the similarity of the microenvironment between bacterial infection lesions and cancer sites, the targeting and accuracy of traditional microenvironment-responsive nanocarriers are questionable. Therefore, finding new specific targets has become a new development direction of nanocarriers in bacterial prevention and treatment. This article reviews the infectious microenvironment induced by bacteria and a series of virulence factors of common pathogenic bacteria and their physiological functions, which may be used as potential targets to improve the targeting accuracy of nanocarriers in lesions.
Collapse
Affiliation(s)
- Ming Jin
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China; Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China
| | - Bin He
- Institute of Animal Husbandry and Veterinary, Wuhan Academy of Agricultural Sciences, China
| | - Xiaoli Cai
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China; Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China
| | - Zhixin Lei
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China; Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China.
| | - Taolei Sun
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China; Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China.
| |
Collapse
|
8
|
Han H, Li S, Xu M, Zhong Y, Fan W, Xu J, Zhou T, Ji J, Ye J, Yao K. Polymer- and lipid-based nanocarriers for ocular drug delivery: Current status and future perspectives. Adv Drug Deliv Rev 2023; 196:114770. [PMID: 36894134 DOI: 10.1016/j.addr.2023.114770] [Citation(s) in RCA: 79] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/21/2023] [Accepted: 03/02/2023] [Indexed: 03/09/2023]
Abstract
Ocular diseases seriously affect patients' vision and life quality, with a global morbidity of over 43 million blindness. However, efficient drug delivery to treat ocular diseases, particularly intraocular disorders, remains a huge challenge due to multiple ocular barriers that significantly affect the ultimate therapeutic efficacy of drugs. Recent advances in nanocarrier technology offer a promising opportunity to overcome these barriers by providing enhanced penetration, increased retention, improved solubility, reduced toxicity, prolonged release, and targeted delivery of the loaded drug to the eyes. This review primarily provides an overview of the progress and contemporary applications of nanocarriers, mainly polymer- and lipid-based nanocarriers, in treating various eye diseases, highlighting their value in achieving efficient ocular drug delivery. Additionally, the review covers the ocular barriers and administration routes, as well as the prospective future developments and challenges in the field of nanocarriers for treating ocular diseases.
Collapse
Affiliation(s)
- Haijie Han
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou 310009, People's Republic of China; Zhejiang Provincial Key Lab of Ophthalmology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou 310009, People's Republic of China
| | - Su Li
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou 310009, People's Republic of China
| | - Mingyu Xu
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou 310009, People's Republic of China
| | - Yueyang Zhong
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou 310009, People's Republic of China; Zhejiang Provincial Key Lab of Ophthalmology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou 310009, People's Republic of China
| | - Wenjie Fan
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou 310009, People's Republic of China
| | - Jingwei Xu
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou 310009, People's Republic of China; Zhejiang Provincial Key Lab of Ophthalmology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou 310009, People's Republic of China
| | - Tinglian Zhou
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou 310009, People's Republic of China
| | - Jian Ji
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, People's Republic of China
| | - Juan Ye
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou 310009, People's Republic of China; Zhejiang Provincial Key Lab of Ophthalmology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou 310009, People's Republic of China.
| | - Ke Yao
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou 310009, People's Republic of China; Zhejiang Provincial Key Lab of Ophthalmology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou 310009, People's Republic of China.
| |
Collapse
|
9
|
Quílez-Alburquerque J, Saad MA, Descalzo AB, Orellana G, Hasan T. Hyaluronic acid-poly(lactic-co-glycolic acid) nanoparticles with a ruthenium photosensitizer cargo for photokilling of oral cancer cells. J Photochem Photobiol A Chem 2023. [DOI: 10.1016/j.jphotochem.2022.114349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
10
|
Zhan L, Tian X, Lin J, Peng X, Zhao G. Glabridin Inhibits Aspergillus fumigatus Growth and Alleviate Inflammation Mediated by Dectin-2 and NLRP3 Inflammasome. Curr Eye Res 2023; 48:348-356. [PMID: 36785524 DOI: 10.1080/02713683.2022.2164779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023]
Abstract
PURPOSE The research was used to uncover the mechanism of glabridin in Aspergillus fumigatus keratitis in anti-fungus and anti-inflammation. METHODS In vitro, RAW 264.7 cells were infected with A. fumigatus with incubation of glabridin in different concentrations. Real-time quantitative polymerase chain reaction (RT‑qPCR), Western blot, and enzyme-linked immunosorbent assay (ELISA) were used to assess the inflammatory severe and alternation with the intervention of Dectin-2 siRNA and glabridin. In vivo, A. fumigatus keratitis mouse models were established by spore intra-stromal injection and treated with glabridin or PBS. And disease scores, inflammatory mediators, and periodic acid-schiff (PAS) staining were exhibited to demonstrate the therapeutic efficiency of glabridin in vivo. Morphological interference assay monitored fungal germination. Scanning and transmission electron microscopy were used to observe the growth of fungi. RESULTS In RAW 264.7 cells and mouse keratitis models, noncytotoxic 16 μg/mL glabridin showed significant inhibition in the expression of Dectin-2, NLRP3, Caspase-1, IL-1β, and TNF-α after A. fumigatus infection, almost similar to the intervention of Dectin-2 siRNA. PAS staining illustrated the reduced hyphal distribution in cornea stroma with glabridin treatment. Glabridin remarkably inhibited A. fumigatus growth through delaying germination and disrupting the integrity of the hyphae membrane. CONCLUSION Glabridin plays an anti-inflammatory role in A. fumigatus challenge via suppression of the Dectin-2 and NLRP3 inflammasome, and plays an anti-fungal role through delaying germination and changing the hyphal integrity.KEY MESSAGESGlabridin plays an anti-inflammatory role in A. fumigatus infection of RAW264.7 cells in a concentration-dependent manner and through Dectin-2 mediation.Glabridin decreases fungal distribution and inflammation in mouse A. fumigatus keratitis.Glabridin inhibits A. fumigatus growth by delaying germination and disrupting cellular structure in vitro.
Collapse
Affiliation(s)
- Lu Zhan
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xue Tian
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jing Lin
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xudong Peng
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China.,Department of Ophthalmology, University of Washington, Seattle, WA, USA
| | - Guiqiu Zhao
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
11
|
Chen X, Yang R, Shen J, Huang Q, Wu Z. Research Progress of Bioinspired Nanostructured Systems for the Treatment of Ocular Disorders. Pharmaceuticals (Basel) 2023; 16:ph16010096. [PMID: 36678597 PMCID: PMC9865244 DOI: 10.3390/ph16010096] [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: 12/14/2022] [Revised: 12/30/2022] [Accepted: 01/06/2023] [Indexed: 01/12/2023] Open
Abstract
How to enhance the bioavailability and prolong the residence time of drugs in the eye present the major barriers to traditional eye delivery. Nanotechnology has been widely used in ocular drug delivery systems because of its advantages of minimizing adverse reactions, decreasing the frequency of administration, prolonging the release time, and improving the bioavailability of the drug in the eye. As natural product-based nanostructured systems, bioinspired nanostructured systems have presented as less toxic, easy to prepare, and cost-effective and have potential application value in the field of nanotechnology. A systematic classification of bioinspired nanostructured systems based on their inspiration source and formulation and their brief applications in disease are presented here. A review of recent research progress of the bioinspired nanostructured systems for the treatment of the anterior and posterior segment of ocular disorders is then presented in detail. Finally, current challenges and future directions with regard to manufacturing bioinspired nanomaterials are provided.
Collapse
Affiliation(s)
- Xuan Chen
- Department of Ophthalmology, Wuxi Second People’s Hospital, Nanjing Medical University, Wuxi 214002, China
| | - Rui Yang
- Research Institute for Reproductive Health and Genetic Diseases, Wuxi Maternity and Child Health Care Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi 214002, China
- Correspondence: (R.Y.); (Z.W.)
| | - Jinyan Shen
- Department of Ophthalmology, Wuxi Second People’s Hospital, Nanjing Medical University, Wuxi 214002, China
| | - Qingyu Huang
- Department of Ophthalmology, Wuxi Second People’s Hospital, Nanjing Medical University, Wuxi 214002, China
| | - Zhifeng Wu
- Department of Ophthalmology, Wuxi Second People’s Hospital, Nanjing Medical University, Wuxi 214002, China
- Department of Ophthalmology, Affiliated Wuxi Clinical College of Nantong University, Wuxi 214002, China
- Correspondence: (R.Y.); (Z.W.)
| |
Collapse
|
12
|
Fang L, Liu J, Liu Z, Zhou H. Immune modulating nanoparticles for the treatment of ocular diseases. J Nanobiotechnology 2022; 20:496. [DOI: 10.1186/s12951-022-01658-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 10/03/2022] [Indexed: 11/25/2022] Open
Abstract
AbstractOcular diseases are increasingly influencing people’s quality of life. Complicated inflammatory mechanisms involved in the pathogenic process of ocular diseases make inflammation-targeting treatment a potential therapeutic approach. The limited efficacy of conventional anti-inflammatory therapeutic strategies, caused by various objective factors, such as complex ocular biological barriers, and subjective factors, such as poor compliance, are promoting the development of new therapeutic methods. With the advantages of considerable tissue permeability, a controllable drug release rate, and selective tissue targeting ability, nanoparticles have successfully captured researchers’ attention and have become a research hotspot in treating ocular diseases. This review will focus on the advantages of nanosystems over traditional therapy, the anti-inflammation mechanisms of nanoparticles, and the anti-inflammatory applications of nanoparticles in different ocular diseases (ocular surface diseases, vitreoretinopathy, uveal diseases, glaucoma, and visual pathway diseases). Furthermore, by analyzing the current situation of nanotherapy and the challenges encountered, we hope to inspire new ideas and incentives for designing nanoparticles more consistent with human physiological characteristics to make progress based on conventional treatments. Overall, some progress has been made in nanoparticles for the treatment of ocular diseases, and nanoparticles have rather broad future clinical translation prospects.
Collapse
|
13
|
Polat HK, Kurt N, Aytekin E, Bozdağ Pehlivan S, Çalış S. Novel Drug Delivery Systems to Improve the Treatment of Keratitis. J Ocul Pharmacol Ther 2022; 38:376-395. [PMID: 35763406 DOI: 10.1089/jop.2021.0127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Keratitis is a disease characterized by inflammation of the cornea caused by different pathogens. It can cause serious visual morbidity if not treated quickly. Depending on the pathogen causing keratitis, eye drops containing antibacterial, antifungal, or antiviral agents such as besiloxacin, moxifloxacin, ofloxacin, voriconazol, econazole, fluconazole, and acyclovir are used, and these drops need to be applied frequently due to their low bioavailability. Studies are carried out on formulations with extended residence time in the cornea and increased permeability. These formulations include various new drug delivery systems such as inserts, nanoparticles, liposomes, niosomes, cubosomes, microemulsions, in situ gels, contact lenses, nanostructured lipid carriers, carbon quantum dots, and microneedles. Ex vivo and in vivo studies with these formulations have shown that the residence time of the active substances in the cornea is prolonged, and their ocular bioavailability is increased. In addition, in vivo studies have shown that these formulations successfully treat keratitis. However, it has been observed that fluoroquinolones are used in most of the studies; similar drug delivery systems are generally preferred for antifungal drugs, and studies for viral and acanthameba keratitis are limited. There is a need for new studies on different types of keratitis and different drug active substances. At the same time, proving the efficacy of drug delivery systems, which give promising results in in vivo animal models, with clinical studies is of great importance for progress in the treatment of keratitis.
Collapse
Affiliation(s)
- Heybet Kerem Polat
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey.,Department of Pharmaceutical Technology, Faculty of Pharmacy, Erzincan Binali Yıldırım University, Erzincan, Turkey
| | - Nihat Kurt
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey.,Department of Pharmaceutical Technology, Faculty of Pharmacy, Tokat Gaziosmanpaşa University, Tokat, Turkey
| | - Eren Aytekin
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Sibel Bozdağ Pehlivan
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Sema Çalış
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| |
Collapse
|
14
|
Gu L, Li C, Lin J, Wang Q, Yin M, Zhang L, Li N, Lin H, You Z, Wang S, Li D, Zhao G. Drug-loaded mesoporous carbon with sustained drug release capacity and enhanced antifungal activity to treat fungal keratitis. BIOMATERIALS ADVANCES 2022; 136:212771. [PMID: 35929310 DOI: 10.1016/j.bioadv.2022.212771] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 03/11/2022] [Accepted: 03/18/2022] [Indexed: 06/15/2023]
Abstract
Fungal keratitis is a severe infectious corneal disease with a high rate of incidence and blindness. Since traditional treatments natamycin (NATA) eye drops, exhibit poor dissolution and bioavailability, and the efficacy of current therapeutic approaches remains limited. In this study, we innovatively utilized mesoporous carbon (Meso-C) and microporous carbon (Micro-C) as nanocarriers loaded with the antifungal drug NATA and silver nanoparticles (Ag-NPs). Porous carbon loaded with NATA and Ag-NPs has not previously been studied in fungal keratitis. Due to the mesoporous structure, high surface area and larger pore volume of Meso-C, it displayed greater superiority in sustained drug release and drug dispersity than Micro-C. Moreover, Meso-C could adsorb inflammatory cytokines during fungal infection. In vitro, Meso-C/NATA/Ag showed excellent antifungal effects. In vivo, compared with pure NATA treatment, Meso-C/NATA/Ag exhibited significantly improved therapeutic effects and reduced dosing frequency when treating fungal keratitis. Our study is the first to report the sustained drug release and improved drug dispersity of Meso-C/NATA and demonstrates that NATA and Ag-NPs-loaded Meso-C has therapeutic effects against fungal keratitis.
Collapse
Affiliation(s)
- Lingwen Gu
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao 266003, PR China
| | - Cui Li
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao 266003, PR China.
| | - Jing Lin
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao 266003, PR China
| | - Qian Wang
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao 266003, PR China
| | - Min Yin
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao 266003, PR China
| | - Lina Zhang
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao 266003, PR China
| | - Na Li
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao 266003, PR China
| | - Hao Lin
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao 266003, PR China
| | - Zhihu You
- State Key Laboratory of Bio-fibers and Eco-textiles, Institute of Marine Biobased Materials, College of Materials Science and Engineering, Qingdao University, Qingdao 266071, PR China
| | - Siyu Wang
- State Key Laboratory of Bio-fibers and Eco-textiles, Institute of Marine Biobased Materials, College of Materials Science and Engineering, Qingdao University, Qingdao 266071, PR China
| | - Daohao Li
- State Key Laboratory of Bio-fibers and Eco-textiles, Institute of Marine Biobased Materials, College of Materials Science and Engineering, Qingdao University, Qingdao 266071, PR China.
| | - Guiqiu Zhao
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao 266003, PR China.
| |
Collapse
|
15
|
Jadi PK, Sharma P, Bhogapurapu B, Roy S. Alternative Therapeutic Interventions: Antimicrobial Peptides and Small Molecules to Treat Microbial Keratitis. Front Chem 2021; 9:694998. [PMID: 34458234 PMCID: PMC8386189 DOI: 10.3389/fchem.2021.694998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 07/02/2021] [Indexed: 01/10/2023] Open
Abstract
Microbial keratitis is a leading cause of blindness worldwide and results in unilateral vision loss in an estimated 2 million people per year. Bacteria and fungus are two main etiological agents that cause corneal ulcers. Although antibiotics and antifungals are commonly used to treat corneal infections, a clear trend with increasing resistance to these antimicrobials is emerging at rapid pace. Extensive research has been carried out to determine alternative therapeutic interventions, and antimicrobial peptides (AMPs) are increasingly recognized for their clinical potential in treating infections. Small molecules targeted against virulence factors of the pathogens and natural compounds are also explored to meet the challenges and growing demand for therapeutic agents. Here we review the potential of AMPs, small molecules, and natural compounds as alternative therapeutic interventions for the treatment of corneal infections to combat antimicrobial resistance. Additionally, we have also discussed about the different formats of drug delivery systems for optimal administration of drugs to treat microbial keratitis.
Collapse
Affiliation(s)
- Praveen Kumar Jadi
- Prof, Brien Holden Eye Research Centre, LV Prasad Eye Institute, Hyderabad, India
| | - Prerana Sharma
- Prof, Brien Holden Eye Research Centre, LV Prasad Eye Institute, Hyderabad, India
- Department of Animal Sciences, University of Hyderabad, Hyderabad, India
| | - Bharathi Bhogapurapu
- Prof, Brien Holden Eye Research Centre, LV Prasad Eye Institute, Hyderabad, India
| | - Sanhita Roy
- Prof, Brien Holden Eye Research Centre, LV Prasad Eye Institute, Hyderabad, India
| |
Collapse
|
16
|
Nguyen DD, Lue SJ, Lai JY. Tailoring therapeutic properties of silver nanoparticles for effective bacterial keratitis treatment. Colloids Surf B Biointerfaces 2021; 205:111856. [PMID: 34022702 DOI: 10.1016/j.colsurfb.2021.111856] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 04/17/2021] [Accepted: 05/13/2021] [Indexed: 11/28/2022]
Abstract
The formulation of nanoparticles with intrinsically therapeutic properties in a tailorable and appropriate manner is critical in nanomedicine for effective treatments of infectious diseases. Here, we present a biomedical strategy to formulate silver nanoparticles (AgNPs) as intrinsically therapeutic agents for the treatment of Staphylococcus aureus (S. aureus) keratitis. Specifically, AgNPs are controllably obtained as spheres, wrapped with a biopolymer, and varied in sizes. in vitro and in vivo studies indicate that biological interactions between the AgNPs and corneal keratocytes, S. aureus bacteria, and blood vessels are strongly determined by the particle sizes. As the size increased from 3.3 ± 0.7 to 37.2 ± 5.3 nm, the AgNPs exhibit better ocular biocompatibility and stronger antiangiogenic activity, but poorer bactericidal performance. In a rabbit model of S. Aureus-induced keratitis, intrastromal injection of AgNP formulations (single dose) show substantial influences of particle size on the treatment efficacy. As the trade-off, AgNPs with medium size of 15.0 ± 3.6 nm reveal as the best therapeutic agent that could offer ∼5.6 and ∼9.1-fold greater corneal thickness recovery respectively compared to those with smaller and larger sizes at 3 days post-administration. These findings suggest an important advance in structural design for formulating intrinsically therapeutic nano-agents toward the efficient management of infectious diseases.
Collapse
Affiliation(s)
- Duc Dung Nguyen
- Graduate Institute of Biomedical Engineering, Chang Gung University, Taoyuan, 33302, Taiwan
| | - Shingjiang Jessie Lue
- Department of Chemical and Materials Engineering, Chang Gung University, Taoyuan, 33302, Taiwan; Division of Joint Reconstruction, Department of Orthopedics, Chang Gung Memorial Hospital, Taoyuan, 33305, Taiwan; Department of Safety, Health and Environmental Engineering, Ming Chi University of Technology, New Taipei City, 24301, Taiwan
| | - Jui-Yang Lai
- Graduate Institute of Biomedical Engineering, Chang Gung University, Taoyuan, 33302, Taiwan; Department of Ophthalmology, Chang Gung Memorial Hospital, Linkou, Taoyuan, 33305, Taiwan; Department of Materials Engineering, Ming Chi University of Technology, New Taipei City, 24301, Taiwan; Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, 33303, Taiwan.
| |
Collapse
|
17
|
Ahamad N, Kar A, Mehta S, Dewani M, Ravichandran V, Bhardwaj P, Sharma S, Banerjee R. Immunomodulatory nanosystems for treating inflammatory diseases. Biomaterials 2021; 274:120875. [PMID: 34010755 DOI: 10.1016/j.biomaterials.2021.120875] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 04/26/2021] [Accepted: 05/02/2021] [Indexed: 02/07/2023]
Abstract
Inflammatory disease (ID) is an umbrella term encompassing all illnesses involving chronic inflammation as the central manifestation of pathogenesis. These include, inflammatory bowel diseases, hepatitis, pulmonary disorders, atherosclerosis, myocardial infarction, pancreatitis, arthritis, periodontitis, psoriasis. The IDs create a severe burden on healthcare and significantly impact the global socio-economic balance. Unfortunately, the standard therapies that rely on a combination of anti-inflammatory and immunosuppressive agents are palliative and provide only short-term relief. In contrast, the emerging concept of immunomodulatory nanosystems (IMNs) has the potential to address the underlying causes and prevent reoccurrence, thereby, creating new opportunities for treating IDs. The IMNs offer exquisite ability to precisely modulate the immune system for a therapeutic advantage. The nano-sized dimension of IMNs allows them to efficiently infiltrate lymphatic drainage, interact with immune cells, and subsequently to undergo rapid endocytosis by hyperactive immune cells (HICs) at inflamed sites. Thus, IMNs serve to restore dysfunctional or HICs and alleviate the inflammation. We identified that different IMNs exert their immunomodulatory action via either of the seven mechanisms to modulate; cytokine production, cytokine neutralization, cellular infiltration, macrophage polarization, HICs growth inhibition, stimulating T-reg mediated tolerance and modulating oxidative-stress. In this article, we discussed representative examples of IMNs by highlighting their rationalization, design principle, and mechanism of action in context of treating various IDs. Lastly, we highlighted technical challenges in the application of IMNs and explored the future direction of research, which could potentially help to overcome those challenges.
Collapse
Affiliation(s)
- Nadim Ahamad
- Nanomedicine Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, 400076, India
| | - Abhinanda Kar
- Nanomedicine Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, 400076, India
| | - Sourabh Mehta
- Nanomedicine Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, 400076, India; IITB-Monash Research Academy IIT Bombay, Powai, Mumbai, 400076, India
| | - Mahima Dewani
- Nanomedicine Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, 400076, India
| | - Vasanthan Ravichandran
- Nanomedicine Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, 400076, India
| | - Prateek Bhardwaj
- Nanomedicine Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, 400076, India
| | - Shivam Sharma
- Nanomedicine Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, 400076, India
| | - Rinti Banerjee
- Nanomedicine Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, 400076, India.
| |
Collapse
|
18
|
Zhang Y, Yu Y, Li G, Zhang X, Wu Z, Lin L. Epithelium-Penetrable Nanoplatform with Enhanced Antibiotic Internalization for Management of Bacterial Keratitis. Biomacromolecules 2021; 22:2020-2032. [PMID: 33880923 DOI: 10.1021/acs.biomac.1c00139] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
A standardized regimen for addressing the adverse effects of bacterial keratitis on vision remains an intractable challenge due to poor epithelial penetration and a short corneal retention time. In this study, a new strategy is proposed to implement the direct transport of antibiotics to bacteria-infected corneas via topical administration of an epithelium-penetrable biodriven nanoplatform, thereby enabling the efficacious treatment of bacterial keratitis. The nanoplatforms were composed of amphiphilic glycopolymers containing boron dipyrromethene and boronic acid moieties with stable fluorescence characteristics and the ability to potentiate epithelial penetration deep into the cornea. The boronic acid-derived nanoplatforms enabled efficient cellular internalization through the high affinity of boric acid groups for the diol-containing bacterial cell wall, resulting in enhanced drug penetration and retention inside the pathogenic bacteria. The bacterial cells formed agglomerations after incorporating the nanoplatforms along with a special mechanism to release the encapsulated cargo in response to in situ bacteria. Compared with the drug alone, this smart system achieved enhanced bacterial mortality and attenuated inflammation associated with Staphylococcus aureus-induced keratitis in rats, demonstrating a paradigm for targeted ocular drug delivery and an alternative strategy for managing bacterial keratitis or other bacterial infections by heightening corneal permeability and transcorneal bioavailability.
Collapse
Affiliation(s)
- Yanlong Zhang
- State Key Laboratory of Precision Measurement Technology and Instrument, School of Precision Instruments & Opto-Electronics Engineering, Tianjin University, Tianjin 300072, China.,Tianjin Key Laboratory of Biomedical Detection Techniques & Instruments, Tianjin University, Tianjin 300072, China.,Tianjin International Joint Research and Development Centre of Ophthalmology and Vision Science, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin 300384, China
| | - Yunjian Yu
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Gang Li
- State Key Laboratory of Precision Measurement Technology and Instrument, School of Precision Instruments & Opto-Electronics Engineering, Tianjin University, Tianjin 300072, China.,Tianjin Key Laboratory of Biomedical Detection Techniques & Instruments, Tianjin University, Tianjin 300072, China
| | - Xinge Zhang
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Zhongming Wu
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China
| | - Ling Lin
- State Key Laboratory of Precision Measurement Technology and Instrument, School of Precision Instruments & Opto-Electronics Engineering, Tianjin University, Tianjin 300072, China.,Tianjin Key Laboratory of Biomedical Detection Techniques & Instruments, Tianjin University, Tianjin 300072, China
| |
Collapse
|
19
|
Antimicrobial nanomedicine for ocular bacterial and fungal infection. Drug Deliv Transl Res 2021; 11:1352-1375. [PMID: 33840082 DOI: 10.1007/s13346-021-00966-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/17/2021] [Indexed: 12/13/2022]
Abstract
Ocular infection induced by bacteria and fungi is a major cause of visual impairment and blindness. Topical administration of antibiotics remains the first-line treatment, as effective eradication of pathogens is the core of the anti-infection strategy. Whereas, eye drops lack efficiency and have relatively low bioavailability. Intraocular injection may cause concurrent ocular damage and secondary infection. In addition, antibiotic-based management can be limited by the low sensitivity to multidrug-resistant bacteria. Nanomedicine is proposed as a prospective, effective, and noninvasive platform to mediate ocular delivery and combat pathogen or even resistant strains. Nanomedicine can not only carry antimicrobial agents to fight against pathogens but also directly active microbicidal capability, killing pathogens. More importantly, by modification, nanomedicine can achieve enhanced residence time and release time on the cornea, and easy penetration through corneal tissues into anterior and posterior segments of the eye, thus improving the therapeutic effect for ocular infection. In this review, several categories of antimicrobial nanomedicine are systematically discussed, where the efficiency and possibility of further embellishment and improvement to adapt to clinical use are also investigated. All in all, novel antimicrobial nanomedicine provides potent and prospective ways to manage severe and refractory ocular infections.
Collapse
|
20
|
Nami S, Aghebati-Maleki A, Aghebati-Maleki L. Current applications and prospects of nanoparticles for antifungal drug delivery. EXCLI JOURNAL 2021; 20:562-584. [PMID: 33883983 PMCID: PMC8056051 DOI: 10.17179/excli2020-3068] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 02/15/2021] [Indexed: 02/06/2023]
Abstract
Currently, the significance of fungi as human pathogens is not medically concealed in the world. Consequently, suitable recognition and treatment of such infections are of great importance and necessitate the need for comprehensive information in this regard. The introduction of new antifungals and their use today, especially in the last two decades, have revolutionized the treatment of fungal infections. On the other hand, increasing drug resistance in the world has overshadowed such developments. The use of NPs results in the treatment of fungal infections and owing to their specific properties, these particles, unlike the pure antibiotics, can exert a greater inhibitory power although with less concentration compared with conventional drugs. Important reasons that have led to the use of antifungal drugs in delivery systems include reduced drug efficacy, limited penetration through tissue, poor aqueous solubility, decreased bioavailability, and poor drug pharmacokinetics. It is therefore hoped that unfavorable properties of antifungal drugs be mitigated via their incorporation into different types of NPs. This review summarizes the different types of NPs as delivery systems of antifungal as well as their advantages over pure drugs.
Collapse
Affiliation(s)
- Sanam Nami
- Department of Parasitology and Mycology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Aghebati-Maleki
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Leili Aghebati-Maleki
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
21
|
Nucleosomal association and altered interactome underlie the mechanism of cataract caused by the R54C mutation of αA-crystallin. Biochim Biophys Acta Gen Subj 2021; 1865:129846. [PMID: 33444727 DOI: 10.1016/j.bbagen.2021.129846] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 01/05/2021] [Accepted: 01/08/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND αA-crystallin plays an important role in eye lens development. Its N-terminal domain is implicated in several important biological functions. Mutations in certain conserved arginine residues in the N-terminal region of αA-crystallin lead to cataract with characteristic cytoplasmic/nuclear aggregation of the mutant protein. In this study, we attempt to gain mechanistic insights into the congenital cataract caused by the R54C mutation in human αA-crystallin. METHODS We used several spectroscopic techniques to investigate the structure and function of the wild-type and R54CαA-crystallin. Immunoprecipitation, chromatin-enrichment followed by western blotting, immunofluorescence and cell-viability assay were performed to study the interaction partners, chromatin-association, stress-like response and cell-death caused by the mutant. RESULTS Although R54CαA-crystallin exhibited slight changes in quaternary structure, its chaperone-like activity was comparable to that of wild-type. When expressed in lens epithelial cells, R54CαA-crystallin exhibited a speckled appearance in the nucleus rather than cytoplasmic localization. R54CαA-crystallin triggered a stress-like response, resulting in nuclear translocation of αB-crystallin, disassembly of cytoskeletal elements and activation of caspase 3, leading to apoptosis. Analysis of the "interactome" revealed an increase in interaction of the mutant protein with nucleosomal histones, and its association with chromatin. CONCLUSIONS The study shows that alteration of "interactome" and nucleosomal association, rather than loss of chaperone-like activity, is the molecular basis of cataract caused by the R54C mutation in αA-crystallin. GENERAL SIGNIFICANCE The study provides a novel mechanism of cataract caused by a mutant of αA-crystallin, and sheds light on the possible mechanism of stress and cell death caused by such nuclear inclusions.
Collapse
|
22
|
Experimental Models for Fungal Keratitis: An Overview of Principles and Protocols. Cells 2020; 9:cells9071713. [PMID: 32708830 PMCID: PMC7408389 DOI: 10.3390/cells9071713] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/08/2020] [Accepted: 07/09/2020] [Indexed: 12/21/2022] Open
Abstract
Fungal keratitis is a potentially blinding infection of the cornea that afflicts diverse patient populations worldwide. The development of better treatment options requires a more thorough understanding of both microbial and host determinants of pathology, and a spectrum of experimental models have been developed toward this end. In vivo (animal) models most accurately capture complex pathological outcomes, but protocols may be challenging to implement and vary widely across research groups. In vitro models allow for the molecular dissection of specific host cell–fungal interactions, but they do so without the appropriate environmental/structural context; ex vivo (corneal explant) models provide the benefits of intact corneal tissue, but they do not provide certain pathological features, such as inflammation. In this review, we endeavor to outline the key features of these experimental models as well as describe key technical variations that could impact study design and outcomes.
Collapse
|
23
|
Sahay P, Singhal D, Nagpal R, Maharana PK, Farid M, Gelman R, Sinha R, Agarwal T, Titiyal JS, Sharma N. Pharmacologic therapy of mycotic keratitis. Surv Ophthalmol 2019; 64:380-400. [DOI: 10.1016/j.survophthal.2019.02.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Revised: 02/06/2019] [Accepted: 02/06/2019] [Indexed: 11/28/2022]
|
24
|
Alvarez-Lorenzo C, Anguiano-Igea S, Varela-García A, Vivero-Lopez M, Concheiro A. Bioinspired hydrogels for drug-eluting contact lenses. Acta Biomater 2019; 84:49-62. [PMID: 30448434 DOI: 10.1016/j.actbio.2018.11.020] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 10/29/2018] [Accepted: 11/14/2018] [Indexed: 12/14/2022]
Abstract
Efficient ocular drug delivery that can overcome the challenges of topical application has been largely pursued. Contact lenses (CLs) may act as light-transparent cornea/sclera bandages for prolonged drug release towards the post-lens tear fluid, if their composition and inner architecture are fitted to the features of the drug molecules. In this review, first the foundations and advantages of using CLs as ocular drug depots are revisited. Then, pros and cons of common strategies to prepare drug-loaded CLs are analyzed on the basis of recent examples, and finally the main section focuses on bioinspired strategies that can overcome some limitations of current designs. Most bioinspired strategies resemble a reverse engineering process to create artificial receptors for the drug inside the CL network by mimicking the human natural binding site of the drug. Related bioinspired strategies are being also tested for designing CLs that elute comfort ingredients mimicking the blinking-associated renewal of eye mucins. Other bioinspired approaches exploit the natural eye variables as stimuli to trigger drug release or take benefit of bio-glues to specifically bind active components to the CL surface. Overall, biomimicking approaches are being revealed as valuable tools to fit the amounts loaded and the release profiles to the therapeutic demands of each pathology. STATEMENT OF SIGNIFICANCE: Biomimetic and bioinspired strategies are remarkable tools for the optimization of drug delivery systems. Translation of the knowledge about how drugs interact with the natural pharmacological receptor and about components and dynamics of anterior eye segment may shed light on the design criteria for obtaining efficient drug-eluting CLs. Current strategies for endowing CLs with controlled drug release performance still require optimization regarding amount loaded, drug retained in the CL structure during storage, regulation of drug release once applied onto the eye, and maintenance of CL physical properties. All these limitations may be addressed through a variety of recently growing bioinspired approaches, which are expected to pave the way of medicated CLs towards the clinics.
Collapse
Affiliation(s)
- Carmen Alvarez-Lorenzo
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, R+D Pharma Group (GI-1645), Facultad de Farmacia and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain.
| | - Soledad Anguiano-Igea
- HGBeyond Materials Science S.L, Edificio Emprendia, Campus Vida s/n, 15782 Santiago de Compostela, Spain
| | - Angela Varela-García
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, R+D Pharma Group (GI-1645), Facultad de Farmacia and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; HGBeyond Materials Science S.L, Edificio Emprendia, Campus Vida s/n, 15782 Santiago de Compostela, Spain
| | - María Vivero-Lopez
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, R+D Pharma Group (GI-1645), Facultad de Farmacia and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Angel Concheiro
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, R+D Pharma Group (GI-1645), Facultad de Farmacia and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| |
Collapse
|
25
|
Lakhani P, Patil A, Majumdar S. Challenges in the Polyene- and Azole-Based Pharmacotherapy of Ocular Fungal Infections. J Ocul Pharmacol Ther 2018; 35:6-22. [PMID: 30481082 DOI: 10.1089/jop.2018.0089] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Polyenes and azoles constitute 2 major drug classes in the antifungal armamentarium used to treat fungal infections of the eye such as fungal keratitis, endophthalmitis, conjunctivitis, and blepharitis. These classes of drugs have come to occupy an important niche in ophthalmic antifungal therapy due to their broad spectrum of activity against a variety of filamentous and yeast-like fungi. Natamycin suspension (Natacyn®), a polyene antifungal drug, is currently the only US FDA-approved formulation for treating ophthalmic fungal infections, whereas the other polyene and azole antifungals such as amphotericin B, fluconazole, itraconazole, ketoconazole, miconazole, voriconazole, and posaconazole are routinely used off-label in the clinical setting. Despite potent antifungal activity, the clinical utility of these agents in ophthalmic infections has been challenged by their physicochemical properties, the unique ocular anatomy and physiology, selective antifungal activity, ocular and systemic toxicity, emergence of resistance and cross-resistance, and absence of reliable techniques for developing a robust in vitro-in vivo correlation. This review discusses the aforementioned challenges and the common approaches undertaken to circumnavigate the difficulties associated with the polyene- and azole-based pharmacotherapy of ophthalmic fungal infections.
Collapse
Affiliation(s)
- Prit Lakhani
- 1 Department of Pharmaceutics and Drug Delivery, School of Pharmacy, University of Mississippi, Oxford, Mississippi.,2 Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, Oxford, Mississippi
| | - Akash Patil
- 1 Department of Pharmaceutics and Drug Delivery, School of Pharmacy, University of Mississippi, Oxford, Mississippi.,2 Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, Oxford, Mississippi
| | - Soumyajit Majumdar
- 1 Department of Pharmaceutics and Drug Delivery, School of Pharmacy, University of Mississippi, Oxford, Mississippi.,2 Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, Oxford, Mississippi
| |
Collapse
|
26
|
Vicente-Pascual M, Albano A, Solinís MÁ, Serpe L, Rodríguez-Gascón A, Foglietta F, Muntoni E, Torrecilla J, Pozo-Rodríguez AD, Battaglia L. Gene delivery in the cornea: in vitro & ex vivo evaluation of solid lipid nanoparticle-based vectors. Nanomedicine (Lond) 2018; 13:1847-1854. [PMID: 29792369 DOI: 10.2217/nnm-2018-0112] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
AIM Inflammation is a process that underlies sight-threatening ocular surface diseases, and gene supplementation with the plasmid that encodes for p-IL10 will allow the sustained de novo synthesis of the cytokine to occur in corneal cells, and provide a long-term anti-inflammatory effect. This work describes the development of solid lipid nanoparticle systems for the delivery of p-IL10 to transfect the cornea. RESULTS In vitro, vectors showed suitable features as nonviral vectors (size, ζ-potential, DNA binding, protection and release), and they were able to enter and transfect human corneal epithelial cells. Ex vivo, the vectors were found to transfect the epithelium, the stroma and the endothelium in rabbit corneal explants. Distribution of gene expression within the cell layers of the cornea depended on the composition of the four vectors evaluated. CONCLUSION Solid lipid nanoparticle-based vectors are promising gene delivery systems for corneal diseases, including inflammation.
Collapse
Affiliation(s)
- Mónica Vicente-Pascual
- Pharmacokinetic, Nanotechnology & Gene Therapy Group (PharmaNanoGene), Faculty of Pharmacy, Centro de investigación Lascaray ikergunea, University of the Basque Country UPV/EHU, Paseo de la Universidad 7, Vitoria-Gasteiz, Spain
| | - Andrea Albano
- Pharmacokinetic, Nanotechnology & Gene Therapy Group (PharmaNanoGene), Faculty of Pharmacy, Centro de investigación Lascaray ikergunea, University of the Basque Country UPV/EHU, Paseo de la Universidad 7, Vitoria-Gasteiz, Spain.,Università degli Studi di Torino, Dipartimento di Scienza e Tecnologia del Farmaco, via Pietro Giuria 9, Turin, Italy
| | - María Á Solinís
- Pharmacokinetic, Nanotechnology & Gene Therapy Group (PharmaNanoGene), Faculty of Pharmacy, Centro de investigación Lascaray ikergunea, University of the Basque Country UPV/EHU, Paseo de la Universidad 7, Vitoria-Gasteiz, Spain
| | - Loredana Serpe
- Università degli Studi di Torino, Dipartimento di Scienza e Tecnologia del Farmaco, via Pietro Giuria 9, Turin, Italy
| | - Alicia Rodríguez-Gascón
- Pharmacokinetic, Nanotechnology & Gene Therapy Group (PharmaNanoGene), Faculty of Pharmacy, Centro de investigación Lascaray ikergunea, University of the Basque Country UPV/EHU, Paseo de la Universidad 7, Vitoria-Gasteiz, Spain
| | - Federica Foglietta
- Università degli Studi di Torino, Dipartimento di Scienza e Tecnologia del Farmaco, via Pietro Giuria 9, Turin, Italy
| | - Elisabetta Muntoni
- Università degli Studi di Torino, Dipartimento di Scienza e Tecnologia del Farmaco, via Pietro Giuria 9, Turin, Italy
| | - Josune Torrecilla
- Pharmacokinetic, Nanotechnology & Gene Therapy Group (PharmaNanoGene), Faculty of Pharmacy, Centro de investigación Lascaray ikergunea, University of the Basque Country UPV/EHU, Paseo de la Universidad 7, Vitoria-Gasteiz, Spain
| | - Ana Del Pozo-Rodríguez
- Pharmacokinetic, Nanotechnology & Gene Therapy Group (PharmaNanoGene), Faculty of Pharmacy, Centro de investigación Lascaray ikergunea, University of the Basque Country UPV/EHU, Paseo de la Universidad 7, Vitoria-Gasteiz, Spain
| | - Luigi Battaglia
- Università degli Studi di Torino, Dipartimento di Scienza e Tecnologia del Farmaco, via Pietro Giuria 9, Turin, Italy
| |
Collapse
|