1
|
Kim HY, Lee JD, Kim H, Kim Y, Park JJ, Oh SB, Goo H, Cho KJ, Kim KB. Mass spectrometry (MS)-based metabolomics of plasma and urine in dry eye disease (DED)-induced rat model. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2025; 88:122-135. [PMID: 39185961 DOI: 10.1080/15287394.2024.2393770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Dry eye disease (DED) is an ophthalmic disease associated with poor quality and quantity of tears, and the number of patients is steadily increasing. The aim of this study was to determine plasma and urine metabolites obtained from DED scopolamine animal model where dry eye conditions (DRY) are induced. It was also of interest to examine whether DED (scopolamine) rat model was exacerbated by treatment with benzalkonium chloride (BAC). Subsequently, plasma and urine metabolites were analyzed using liquid chromatography (LC) and gas chromatography (GC)-mass spectrometry (MS), respectively. Data demonstrated that DED indicators such as tear volume, tear breakup time (TBUT), and corneal damage in the DED groups (DRY and BAC group) differed from those of control (CON). Similar results were noted in inflammatory factors such as interleukin (IL-1β), IL-6, and tumor necrosis factor (TNF)-α. In the partial least squares-discriminant analysis (PLS-DA) score plots, the three groups were distinctly separated from each other. In addition, the related metabolites were also associated with these distinct separations as evidenced by 9 and 14 in plasma and urine, respectively. Almost all of the selected metabolites were decreased in the DRY group compared to CON, and the BAC group was lower than the DRY. In plasma and urine, lysophosphatidylcholine/lysophosphatidylethanolamine, organic acids, amino acids, and sugars varied between three groups, and these metabolites were related to inflammation and oxidative stress. Data suggest that treatment with scopolamine with/without BAC-induced DED and affected the level of systemic metabolites involved in inflammation and oxidative stress.
Collapse
Affiliation(s)
- Hyang Yeon Kim
- College of Pharmacy, Dankook University, Cheonan, Chungnam, Republic of Korea
- Center for Human Risk Assessment, Dankook University, Cheonan, Chungnam, Republic of Korea
| | - Jung Dae Lee
- College of Pharmacy, Dankook University, Cheonan, Chungnam, Republic of Korea
- Center for Human Risk Assessment, Dankook University, Cheonan, Chungnam, Republic of Korea
| | - HongYoon Kim
- College of Pharmacy, Dankook University, Cheonan, Chungnam, Republic of Korea
- Center for Human Risk Assessment, Dankook University, Cheonan, Chungnam, Republic of Korea
| | - YuJin Kim
- College of Pharmacy, Dankook University, Cheonan, Chungnam, Republic of Korea
- Center for Human Risk Assessment, Dankook University, Cheonan, Chungnam, Republic of Korea
| | - Jin Ju Park
- College of Pharmacy, Dankook University, Cheonan, Chungnam, Republic of Korea
- Center for Human Risk Assessment, Dankook University, Cheonan, Chungnam, Republic of Korea
| | - Soo Bean Oh
- Department of Ophthalmology, College of Medicine, Dankook University, Cheonan, Chungnam, Republic of Korea
| | - Hyeyoon Goo
- Department of Ophthalmology, College of Medicine, Dankook University, Cheonan, Chungnam, Republic of Korea
| | - Kyong Jin Cho
- Department of Ophthalmology, College of Medicine, Dankook University, Cheonan, Chungnam, Republic of Korea
| | - Kyu-Bong Kim
- College of Pharmacy, Dankook University, Cheonan, Chungnam, Republic of Korea
- Center for Human Risk Assessment, Dankook University, Cheonan, Chungnam, Republic of Korea
| |
Collapse
|
2
|
Kim SJ, Guo N, Tan ZY, Gao X, Fattahi P, Liu H, Chang J, Younesi M, Jung S, Chung Y, Song M, Jung M, Huh DD. A Bioengineered Model of the Human Cornea for Preclinical Assessment of Human Ocular Exposure to Environmental Toxicants. Adv Healthc Mater 2025:e2402408. [PMID: 39838783 DOI: 10.1002/adhm.202402408] [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: 07/01/2024] [Revised: 01/02/2025] [Indexed: 01/23/2025]
Abstract
Here a bioengineered platform is introduced to investigate adverse effects of environmental materials on the human cornea. Using primary cells, this system is capable of reproducing the differentiated corneal epithelium and its underlying stroma in the human eye, which can then be treated with externally applied solid, liquid, or gaseous substances in a controlled manner and under physiologically relevant conditions. The proof-of-principle of how this system can be used to simulate human ocular exposure to different classes of environmental toxicants for direct visualization and quantitative analysis of their potential to induce acute corneal injury and inflammation is demonstrated. This model can also be further engineered to create an electromechanically actuated array of multiple human corneal tissues that can emulate spontaneous eye blinking. Using this advanced system, it is shown that blinking-like mechanical motions may play a protective role against adverse effects of environmental toxicants. This work yields an immediately deployable in vitro technology for screening ocular toxicity of existing and emerging environmental materials of various types and may enable the development of more realistic, human-relevant preclinical toxicology models complementary to traditional animal testing.
Collapse
Affiliation(s)
- Se-Jeong Kim
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Ning Guo
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Zong Yao Tan
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Xing Gao
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Pouria Fattahi
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Haijiao Liu
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jeehan Chang
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Mousa Younesi
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Selice Jung
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Yewhan Chung
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Minkyung Song
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Michelle Jung
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Dan Dongeun Huh
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
- NSF Science and Technology Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Center for Innovation and Precision Dentistry, University of Pennsylvania, Philadelphia, PA, 19104, USA
| |
Collapse
|
3
|
Randall Harrell C, Djonov V, Volarevic A, Arsenijevic A, Volarevic V. Mesenchymal Stem Cell-Sourced Exosomes as Potentially Novel Remedies for Severe Dry Eye Disease. J Ophthalmol 2025; 2025:5552374. [PMID: 39839752 PMCID: PMC11748739 DOI: 10.1155/joph/5552374] [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: 03/31/2024] [Revised: 12/21/2024] [Accepted: 01/02/2025] [Indexed: 01/23/2025] Open
Abstract
Severe dry eye disease (DED) is an inflammatory condition characterized by a lack of sufficient moisture or lubrication on the surface of the eye, significantly impacting the quality of life and visual function. Since detrimental immune response is crucially responsible for the development and aggravation of DED, therapeutic agents which modulate phenotype and function of eye-infiltrated inflammatory immune cells could be used for the treatment of severe DED. Due to their potent immunomodulatory properties, mesenchymal stem cells (MSCs) represent potentially new remedies for the treatment of inflammatory eye diseases. The majority of MSC-sourced bioactive factors are contained within MSC-derived exosomes (MSC-Exos), nano-sized extracellular vesicles which, due to their nanosize dimension and lipid envelope, easily by pass all biological barriers in the body and deliver their cargo directly into the target immune cells. MSC-Exos contain a variety of bioactive proteins (growth factors, immunoregulatory molecules, cytokines, and chemokines) lipids, and microRNAs (miRNAs) which affect viability, proliferation, phenotype, and function of eye-infiltrated immune cells. Accordingly, MSC-Exos may modulate the progression of inflammatory eye diseases, including DED. Therefore, in this review article, we summarized the current knowledge regarding molecular and cellular mechanisms which were responsible for trophic, anti-inflammatory, immunoregulatory, and regenerative properties of MSC-Exos in the treatment of severe DED. For this purpose, an extensive literature review was carried out in February 2024 across several databases (Medline, Embase, and Google Scholar), from 2000 to the present. Eligible studies delineated molecular and cellular mechanisms responsible for the MSC-Exos-based modulation of immune cell-driven eye inflammation in DED, and their findings were analyzed in this review. Results obtained in these studies demonstrated beneficial effects of MSC-Exos in the treatment of severe DED, paving the way for their future clinical use in ophthalmology. Trial Registration: ClinicalTrials.gov identifier: NCT04213248, NCT06475027, NCT06543667, NCT05738629.
Collapse
Affiliation(s)
- Carl Randall Harrell
- Department of Molecular Biology, Regenerative Processing Plant, LLC, 34176 US Highway 19 N, Palm Harbor, Florida, USA
| | - Valentin Djonov
- Department of Anatomy, Institute of Anatomy, University of Bern, Baltzerstrasse 2, Bern 3012, Switzerland
| | - Ana Volarevic
- Departments of Psychology, Center for Research on Harmful Effects of Biological and Chemical Hazards, Faculty of Medical Sciences University of Kragujevac, 69 Svetozara Markovica Street, Kragujevac 34000, Serbia
| | - Aleksandar Arsenijevic
- Departments of Genetics, Microbiology and Immunology, Center for Research on Harmful Effects of Biological and Chemical Hazards, Faculty of Medical Sciences University of Kragujevac, 69 Svetozara Markovica Street, Kragujevac 34000, Serbia
| | - Vladislav Volarevic
- Departments of Genetics, Microbiology and Immunology, Center for Research on Harmful Effects of Biological and Chemical Hazards, Faculty of Medical Sciences University of Kragujevac, 69 Svetozara Markovica Street, Kragujevac 34000, Serbia
- Department of Biology, Faculty of Pharmacy Novi Sad, Trg Mladenaca 5, Novi Sad 21000, Serbia
| |
Collapse
|
4
|
Thomazini VC, Ramalho LX, Dias MS, Vieira RC, Reis JJP, Raggi IBM, Ferreira LK, Souza MO, Giuberti CS, Villanova JCO. Excipients of concern in the package inserts of human medicines prescribed in veterinary pharmacotherapy: frequency and implications for animal health. AN ACAD BRAS CIENC 2024; 96:e20240656. [PMID: 39630803 DOI: 10.1590/0001-3765202420240656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Indexed: 12/07/2024] Open
Abstract
The therapeutic arsenal for veterinary pharmacotherapy includes both medicines for exclusive veterinary use and human medicines prescribed off-label for animals. Unlike medicines for exclusive veterinary use, the package inserts of human medicines detail the qualitative composition of the excipients in these products. Some excipients may cause adverse reactions in specific animal populations, necessitating that healthcare professionals identify and avoid administering medicines containing these excipients to susceptible animals. In this study, we analyzed 120 package inserts for human medicines prescribed for animals and, identified 14 excipients of concern for this population: 10 in liquid products, 6 in semi-solid products, and 5 in solid products. The identified excipients include ethyl alcohol, benzyl alcohol, benzalkonium chloride, mannitol, sodium metabisulfite, sodium lauryl sulfate, propylene glycol, polyethylene glycol, polysorbate, and ethoxylated castor oil. Although off-label use of human medicines in veterinary practice is permitted, the study suggests that regulatory agencies and professional councils should raise awareness among veterinary prescribers and pharmacists regarding these excipients. This unprecedented study highlights the urgent need to address this issue, aiming to encourage research on excipient safety in animal populations and strengthen veterinary pharmacovigilance services.
Collapse
Affiliation(s)
- Vanessa C Thomazini
- Universidade Federal do Espírito Santo - UFES, Programa de Pós-graduação em Ciências Veterinárias, Centro de Ciências Agrárias e Engenharias - CCAE, Av. Alto Universitário, s/n, 29500-000 Alegre, ES, Brazil
| | - Letícia X Ramalho
- Universidade Federal do Espírito Santo - UFES, Programa de Pós-graduação em Ciências Veterinárias, Centro de Ciências Agrárias e Engenharias - CCAE, Av. Alto Universitário, s/n, 29500-000 Alegre, ES, Brazil
| | - Marcela S Dias
- Universidade Federal do Espírito Santo - UFES, Centro de Ciências Exatas e da Saúde - CCENS, Laboratório de Desenvolvimento de Produtos Farmacêuticos, Av. Alto Universitário, s/n, 29500-000 Alegre, ES, Brazil
| | - Ramon C Vieira
- Universidade Federal do Espírito Santo - UFES, Centro de Ciências Exatas e da Saúde - CCENS, Laboratório de Desenvolvimento de Produtos Farmacêuticos, Av. Alto Universitário, s/n, 29500-000 Alegre, ES, Brazil
| | - João José P Reis
- Universidade Federal do Espírito Santo - UFES, Centro de Ciências Exatas e da Saúde - CCENS, Laboratório de Desenvolvimento de Produtos Farmacêuticos, Av. Alto Universitário, s/n, 29500-000 Alegre, ES, Brazil
| | - Isabela B M Raggi
- Universidade Federal do Espírito Santo - UFES, Centro de Ciências Exatas e da Saúde - CCENS, Laboratório de Desenvolvimento de Produtos Farmacêuticos, Av. Alto Universitário, s/n, 29500-000 Alegre, ES, Brazil
| | - Larissa Kéllen Ferreira
- Universidade Federal do Espírito Santo - UFES, Centro de Ciências Exatas e da Saúde - CCENS, Laboratório de Desenvolvimento de Produtos Farmacêuticos, Av. Alto Universitário, s/n, 29500-000 Alegre, ES, Brazil
| | - Maurilia O Souza
- Universidade Federal do Espírito Santo - UFES, Programa de Pós-graduação em Ciências Farmacêuticas, Centro de Ciências da Saúde - CCS, Av. Marechal Campos, 1468, 29047-105 Vitória, ES, Brazil
| | - Cristiane S Giuberti
- Universidade Federal do Espírito Santo - UFES, Programa de Pós-graduação em Ciências Farmacêuticas, Centro de Ciências da Saúde - CCS, Av. Marechal Campos, 1468, 29047-105 Vitória, ES, Brazil
| | - Janaína Cecília O Villanova
- Universidade Federal do Espírito Santo - UFES, Programa de Pós-graduação em Ciências Veterinárias, Centro de Ciências Agrárias e Engenharias - CCAE, Av. Alto Universitário, s/n, 29500-000 Alegre, ES, Brazil
- Universidade Federal do Espírito Santo - UFES, Centro de Ciências Exatas e da Saúde - CCENS, Laboratório de Desenvolvimento de Produtos Farmacêuticos, Av. Alto Universitário, s/n, 29500-000 Alegre, ES, Brazil
- Universidade Federal do Espírito Santo - UFES, Programa de Pós-graduação em Ciências Farmacêuticas, Centro de Ciências da Saúde - CCS, Av. Marechal Campos, 1468, 29047-105 Vitória, ES, Brazil
| |
Collapse
|
5
|
Wei W, Cao H, Shen D, Sun X, Jia Z, Zhang M. Antioxidant Carbon Dots Nanozyme Loaded in Thermosensitive in situ Hydrogel System for Efficient Dry Eye Disease Treatment. Int J Nanomedicine 2024; 19:4045-4060. [PMID: 38736656 PMCID: PMC11088389 DOI: 10.2147/ijn.s456613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Accepted: 04/30/2024] [Indexed: 05/14/2024] Open
Abstract
Purpose Dry eye disease (DED) is a multifactorial ocular surface disease with a rising incidence. Therefore, it is urgent to construct a reliable and efficient drug delivery system for DED treatment. Methods In this work, we loaded C-dots nanozyme into a thermosensitive in situ gel to create C-dots@Gel, presenting a promising composite ocular drug delivery system to manage DED. Results This composite ocular drug delivery system (C-dots@Gel) demonstrated the ability to enhance adherence to the corneal surface and extend the ocular surface retention time, thereby enhancing bioavailability. Furthermore, no discernible ocular surface irritation or systemic toxicity was observed. In the DED mouse model induced by benzalkonium chloride (BAC), it was verified that C-dots@Gel effectively mitigated DED by stabilizing the tear film, prolonging tear secretion, repairing corneal surface damage, and augmenting the population of conjunctival goblet cells. Conclusion Compared to conventional dosage forms (C-dots), the C-dots@Gel could prolong exhibited enhanced retention time on the ocular surface and increased bioavailability, resulting in a satisfactory therapeutic outcome for DED.
Collapse
Affiliation(s)
- Wei Wei
- School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, 710061, People’s Republic of China
- Xi’an No.1 Hospital, Shaanxi Institute of Ophthalmology, Shaanxi Key Laboratory of Ophthalmology, Clinical Research Center for Ophthalmology Diseases of Shaanxi Province, First Affiliated Hospital of Northwestern University, Xi’an, Shaanxi, 710002, People’s Republic of China
| | - Haili Cao
- Xi’an No.1 Hospital, Shaanxi Institute of Ophthalmology, Shaanxi Key Laboratory of Ophthalmology, Clinical Research Center for Ophthalmology Diseases of Shaanxi Province, First Affiliated Hospital of Northwestern University, Xi’an, Shaanxi, 710002, People’s Republic of China
| | - Di Shen
- Xi’an No.1 Hospital, Shaanxi Institute of Ophthalmology, Shaanxi Key Laboratory of Ophthalmology, Clinical Research Center for Ophthalmology Diseases of Shaanxi Province, First Affiliated Hospital of Northwestern University, Xi’an, Shaanxi, 710002, People’s Republic of China
| | - Xiyu Sun
- Xi’an No.1 Hospital, Shaanxi Institute of Ophthalmology, Shaanxi Key Laboratory of Ophthalmology, Clinical Research Center for Ophthalmology Diseases of Shaanxi Province, First Affiliated Hospital of Northwestern University, Xi’an, Shaanxi, 710002, People’s Republic of China
| | - Zhenzhen Jia
- School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, 710061, People’s Republic of China
| | - Mingzhen Zhang
- School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, 710061, People’s Republic of China
| |
Collapse
|
6
|
Zhu D, Zheng N, Deng K, Li L. Aurantio-obtusin Alleviates Dry Eye Disease by Targeting NF-κB/NLRP3 Signaling in Rodent Models. Biochem Genet 2024; 62:1-14. [PMID: 37633872 DOI: 10.1007/s10528-023-10471-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 07/24/2023] [Indexed: 08/28/2023]
Abstract
Dry eye disease (DED) is a common inflammatory ocular surface disorder, seriously affecting the quality of life of patients. Aurantio-obtusin (AO) is a bioactive anthraquinone compound isolated from Semen Cassiae which has multiple pharmacological activities. Nonetheless, the specific function of AO in DED is unclarified. In this study, a rodent DED model was established by benzalkonium chloride (BAC) induction, followed by topical administration of AO. The results showed that topical application of AO increased tear production, mitigated ocular surface disruption and maintained the number of goblet cells in BAC-induced DED rats (p˂0.05). ELISA revealed that AO treatment significantly (p˂0.001) reduced the production of proinflammatory cytokines and chemokines in the conjunctiva and cornea of BAC-induced DED rats. Immunohistochemical staining and western blotting showed that AO treatment suppressed the expression levels of NLR family pyrin domain containing 3 (NLRP3) inflammasome-related proteins, and inhibited activation of nuclear factor kappa B (NF-κB) signaling pathway in rat conjunctiva and cornea (p˂0.001). In conclusion, AO treatment alleviates BAC-induced DED in rats by inhibiting NF-κB/NLRP3 inflammasome signaling pathway.
Collapse
Affiliation(s)
- Dan Zhu
- Department of Ophthalmology, Hubei Provincial Hospital of Traditional Chinese Medicine, 430061, Wuhan, Hubei, China
| | - Na Zheng
- Department of Otolaryngology, Hubei Provincial Hospital of Traditional Chinese Medicine, 430061, Wuhan, Hubei, China
| | - Kebin Deng
- Department of Otolaryngology, Hubei Provincial Hospital of Traditional Chinese Medicine, 430061, Wuhan, Hubei, China
| | - Liangchang Li
- Department of Ophthalmology, Hubei Provincial Hospital of Traditional Chinese Medicine, 430061, Wuhan, Hubei, China.
- Hubei Provincial Hospital of Traditional Chinese Medicine, No. 4, Huayuan Hill, Wuchang District, Wuhan, Hubei, China.
| |
Collapse
|
7
|
Wu CM, Mao JW, Zhu JZ, Xie CC, Yao JY, Yang XQ, Xiang M, He YF, Tong X, Litifu D, Xiong XY, Cheng MN, Zhu FH, He SJ, Lin ZM, Zuo JP. DZ2002 alleviates corneal angiogenesis and inflammation in rodent models of dry eye disease via regulating STAT3-PI3K-Akt-NF-κB pathway. Acta Pharmacol Sin 2024; 45:166-179. [PMID: 37605050 PMCID: PMC10770170 DOI: 10.1038/s41401-023-01146-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 07/25/2023] [Indexed: 08/23/2023] Open
Abstract
Dry eye disease (DED) is a prevalent ocular disorder with a multifactorial etiology. The pre-angiogenic and pre-inflammatory milieu of the ocular surface plays a critical role in its pathogenesis. DZ2002 is a reversible type III S-adenosyl-L-homocysteine hydrolase (SAHH) inhibitor, which has shown excellent anti-inflammatory and immunosuppressive activities in vivo and in vitro. In this study, we evaluated the therapeutic potential of DZ2002 in rodent models of DED. SCOP-induced dry eye models were established in female rats and mice, while BAC-induced dry eye model was established in female rats. DZ2002 was administered as eye drops (0.25%, 1%) four times daily (20 μL per eye) for 7 or 14 consecutive days. We showed that topical application of DZ2002 concentration-dependently reduced corneal neovascularization and corneal opacity, as well as alleviated conjunctival irritation in both DED models. Furthermore, we observed that DZ2002 treatment decreased the expression of genes associated with angiogenesis and the levels of inflammation in the cornea and conjunctiva. Moreover, DZ2002 treatment in the BAC-induced DED model abolished the activation of the STAT3-PI3K-Akt-NF-κB pathways in corneal tissues. We also found that DZ2002 significantly inhibited the proliferation, migration, and tube formation of human umbilical endothelial cells (HUVECs) while downregulating the activation of the STAT3-PI3K-Akt-NF-κB pathway. These results suggest that DZ2002 exerts a therapeutic effect on corneal angiogenesis in DED, potentially by preventing the upregulation of the STAT3-PI3K-Akt-NF-κB pathways. Collectively, DZ2002 is a promising candidate for ophthalmic therapy, particularly in treating DED.
Collapse
Affiliation(s)
- Chun-Mei Wu
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jia-Wen Mao
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jin-Zhi Zhu
- Department of Pharmacy, Shanghai Xuhui Central Hospital, Shanghai, 200031, China
| | - Can-Can Xie
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- Laboratory of Immunology and Virology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Jia-Ying Yao
- College of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330004, China
- Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xiao-Qian Yang
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Mai Xiang
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yi-Fan He
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiao Tong
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Dilinaer Litifu
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiao-Yu Xiong
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Meng-Nan Cheng
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Feng-Hua Zhu
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Shi-Jun He
- Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Ze-Min Lin
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
| | - Jian-Ping Zuo
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
- Laboratory of Immunology and Virology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
8
|
Han R, Gao J, Wang L, Hao P, Chen X, Wang Y, Jiang Z, Jiang L, Wang T, Zhu L, Li X. MicroRNA-146a negatively regulates inflammation via the IRAK1/TRAF6/NF-κB signaling pathway in dry eye. Sci Rep 2023; 13:11192. [PMID: 37433841 DOI: 10.1038/s41598-023-38367-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 07/07/2023] [Indexed: 07/13/2023] Open
Abstract
Inflammation is a key factor in the pathogenesis of dry eye disease (DED). We aimed to investigate the role of microRNA-146a (miR-146a) in regulating corneal inflammation in a mouse model of benzalkonium chloride (BAC)-induced dry eye and the TNF-α-induced NF-κB signaling pathway in human corneal epithelial cells (HCECs). A mouse model of dry eye was established by administering with BAC to BALB/c mice, and the expression of TNF-α, IL-1β, IL-6, IL-8, cyclooxygenase 2 (COX2), interleukin-1 receptor-associated kinase 1 (IRAK1) and TNF receptor-associated factor 6 (TRAF6) in the corneas of dry eye model mice was significantly increased; this was accompanied by the upregulation of miR-146a and activation of the NF-κB pathway. In vitro, TNF-α induced miR-146a expression in HCECs, while the NF-κB inhibitor SC-514 reduced the expression of miR-146a. Overexpression of miR-146a decreased the expression of IRAK1 and TRAF6, which have been identified as targets of miR-146a. Furthermore, overexpression of miR-146a suppressed NF-κB p65 translocation from the cytoplasm to the nucleus. Moreover, overexpression of miR-146a attenuated the TNF-α-induced expression of IL-6, IL-8, COX2 and intercellular adhesion molecule 1 (ICAM1), while inhibition of miR-146a exerted the opposite effect. Our results suggest that miR-146a mediates the inflammatory response in DED. MiR-146a negatively regulates inflammation in HCECs through the IRAK1/TRAF6/NF-κB pathway, and this may serve as a potential therapeutic approach for the treatment of DED.
Collapse
Affiliation(s)
- Ruifang Han
- Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Hospital, Tianjin Eye Institute, Nankai University Affiliated Eye Hospital, Clinical College of Ophthalmology of Tianjin Medical University, No.4 Gansu Road, Heping District, Tianjin, 300020, China
| | - Juan Gao
- Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Hospital, Tianjin Eye Institute, Nankai University Affiliated Eye Hospital, Clinical College of Ophthalmology of Tianjin Medical University, No.4 Gansu Road, Heping District, Tianjin, 300020, China
| | - Liming Wang
- Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Hospital, Tianjin Eye Institute, Nankai University Affiliated Eye Hospital, Clinical College of Ophthalmology of Tianjin Medical University, No.4 Gansu Road, Heping District, Tianjin, 300020, China
| | - Peng Hao
- Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Hospital, Tianjin Eye Institute, Nankai University Affiliated Eye Hospital, Clinical College of Ophthalmology of Tianjin Medical University, No.4 Gansu Road, Heping District, Tianjin, 300020, China
| | - Xi Chen
- Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Hospital, Tianjin Eye Institute, Nankai University Affiliated Eye Hospital, Clinical College of Ophthalmology of Tianjin Medical University, No.4 Gansu Road, Heping District, Tianjin, 300020, China
| | - Yuchuan Wang
- Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Hospital, Tianjin Eye Institute, Nankai University Affiliated Eye Hospital, Clinical College of Ophthalmology of Tianjin Medical University, No.4 Gansu Road, Heping District, Tianjin, 300020, China
| | - Zhixin Jiang
- Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Hospital, Tianjin Eye Institute, Nankai University Affiliated Eye Hospital, Clinical College of Ophthalmology of Tianjin Medical University, No.4 Gansu Road, Heping District, Tianjin, 300020, China
| | - Li Jiang
- Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Hospital, Tianjin Eye Institute, Nankai University Affiliated Eye Hospital, Clinical College of Ophthalmology of Tianjin Medical University, No.4 Gansu Road, Heping District, Tianjin, 300020, China
| | - Ting Wang
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China
| | - Lin Zhu
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China
| | - Xuan Li
- Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Hospital, Tianjin Eye Institute, Nankai University Affiliated Eye Hospital, Clinical College of Ophthalmology of Tianjin Medical University, No.4 Gansu Road, Heping District, Tianjin, 300020, China.
| |
Collapse
|
9
|
Suanno G, Fonteyne P, Ferrari G. Neurosensory abnormalities and stability of a mouse model of dry eye disease. Exp Eye Res 2023; 232:109516. [PMID: 37209768 DOI: 10.1016/j.exer.2023.109516] [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: 02/15/2023] [Revised: 04/05/2023] [Accepted: 05/17/2023] [Indexed: 05/22/2023]
Abstract
This study aimed to use a mouse model of dry eye disease (DED) induced by topical administration of benzalkonium chloride (BAK) and assess its stability and the presence of neurosensory abnormalities, including ocular pain. Eight-week-old C57BL6/6 N male mice were used in this study. Mice were treated with 10 μL of 0.2% BAK dissolved in artificial tears (AT), administered twice daily for 7 days. After one week, animals were randomized into two groups: one was administered with 0.2% BAK in AT once per day for 7 days, while the other was not further treated. Corneal epitheliopathy was quantified at days 0, 3, 7, 12, and 14. Moreover, tear secretions, corneal nociception, and corneal nerve integrity were measured after BAK treatment. After sacrifice, corneas were dissected to assess nerve density and leukocyte infiltration by immunofluorescence. Topical BAK instillation for 14 days significantly increased corneal fluorescein staining (p < 0.0001) compared to day 0. On the other hand, interruption of BAK instillation was associated with improvement of corneal epitheliopathy (day 12, p < 0.0001; day 14, p < 0.001). BAK treatment increased ocular pain (p < 0.0001) and resulted in a significant increase in leukocyte infiltration in the cornea (p < 0.01). Moreover, corneal sensitivity was reduced (p < 0.0001), together with corneal nerve density (p < 0.0001) and tear secretion (p < 0.0001). One week twice a day, followed by one additional week once a day, of 0.2% BAK topical administration induces stable clinical and histological signs of DED, which is associated with neurosensory abnormalities, including pain.
Collapse
Affiliation(s)
- Giuseppe Suanno
- Cornea and Ocular Surface Disease Unit, Eye Repair Lab, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Philippe Fonteyne
- Cornea and Ocular Surface Disease Unit, Eye Repair Lab, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Giulio Ferrari
- Cornea and Ocular Surface Disease Unit, Eye Repair Lab, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy.
| |
Collapse
|
10
|
Chintala SK, Pan J, Satapathy S, Condruti R, Hao Z, Liu PW, O’Conner CF, Barr JT, Wilson MR, Jeong S, Fini ME. Recombinant Human Clusterin Seals Damage to the Ocular Surface Barrier in a Mouse Model of Ophthalmic Preservative-Induced Epitheliopathy. Int J Mol Sci 2023; 24:981. [PMID: 36674497 PMCID: PMC9861099 DOI: 10.3390/ijms24020981] [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: 11/21/2022] [Revised: 12/23/2022] [Accepted: 12/31/2022] [Indexed: 01/07/2023] Open
Abstract
There is a significant unmet need for therapeutics to treat ocular surface barrier damage, also called epitheliopathy, due to dry eye and related diseases. We recently reported that the natural tear glycoprotein CLU (clusterin), a molecular chaperone and matrix metalloproteinase inhibitor, seals and heals epitheliopathy in mice subjected to desiccating stress in a model of aqueous-deficient/evaporative dry eye. Here we investigated CLU sealing using a second model with features of ophthalmic preservative-induced dry eye. The ocular surface was stressed by topical application of the ophthalmic preservative benzalkonium chloride (BAC). Then eyes were treated with CLU and sealing was evaluated immediately by quantification of clinical dye uptake. A commercial recombinant form of human CLU (rhCLU), as well as an rhCLU form produced in our laboratory, designed to be compatible with U.S. Food and Drug Administration guidelines on current Good Manufacturing Practices (cGMP), were as effective as natural plasma-derived human CLU (pCLU) in sealing the damaged ocular surface barrier. In contrast, two other proteins found in tears: TIMP1 and LCN1 (tear lipocalin), exhibited no sealing activity. The efficacy and selectivity of rhCLU for sealing of the damaged ocular surface epithelial barrier suggests that it could be of therapeutic value in treating BAC-induced epitheliopathy and related diseases.
Collapse
Affiliation(s)
- Shravan K. Chintala
- USC Institute for Genetic Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA 90033, USA
| | - Jinhong Pan
- New England Eye Center, Tufts Medical Center, Department of Ophthalmology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Sandeep Satapathy
- School of Chemistry and Molecular Bioscience, Molecular Horizons Research Institute, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Rebecca Condruti
- Training Program in Cell, Molecular and Developmental Biology, Graduate School of Biomedical Sciences, Tufts University, Boston, MA 02111, USA
| | - Zixuan Hao
- Training Program in Pharmacology and Drug Development, Graduate School of Biomedical Sciences, Tufts University, Boston, MA 02111, USA
| | - Pei-wen Liu
- Training Program in Pharmacology and Drug Development, Graduate School of Biomedical Sciences, Tufts University, Boston, MA 02111, USA
| | - Christian F. O’Conner
- Doctor of Medicine Training Program, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Joseph T. Barr
- The Ohio State University College of Optometry, Columbus, OH 43210, USA
| | - Mark R. Wilson
- School of Chemistry and Molecular Bioscience, Molecular Horizons Research Institute, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Shinwu Jeong
- USC Roski Eye Institute, Department of Ophthalmology, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA 90033, USA
| | - M. Elizabeth Fini
- New England Eye Center, Tufts Medical Center, Department of Ophthalmology, Tufts University School of Medicine, Boston, MA 02111, USA
| |
Collapse
|
11
|
Wang HH, Chen WY, Huang YH, Hsu SM, Tsao YP, Hsu YH, Chang MS. Interleukin-20 is involved in dry eye disease and is a potential therapeutic target. J Biomed Sci 2022; 29:36. [PMID: 35681232 PMCID: PMC9178884 DOI: 10.1186/s12929-022-00821-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 05/29/2022] [Indexed: 11/23/2022] Open
Abstract
Background Dry eye disease (DED) is a common disease in ophthalmology, affecting millions of people worldwide. Recent studies have shown that inflammation is the core mechanism of DED. IL-20 is a proinflammatory cytokine involved in various inflammatory diseases. Therefore, we aimed to explore the role of this cytokine in the pathogenesis of DED and evaluate the therapeutic potential of the anti-IL-20 monoclonal antibody (mAb) 7E for DED treatment. Methods Clinical tear samples from patients with DED and non-DED controls were collected and their IL-20 protein levels were determined. We established three DED animal models to explore the role of IL-20 and the efficacy of IL-20 antibody in DED. Benzalkonium chloride (BAC)-induced over-evaporative DED, extra-orbital lacrimal gland excision (LGE)-induced aqueous tear-deficient DED, and desiccating stress (DS)-induced combined over-evaporative and aqueous tear-deficient DED animal models were established to investigate the role of IL-20. The anti-IL-20 antibody 7E was established to neutralize IL-20 activity. The effects of IL-20 or 7E on human corneal epithelial cells and macrophages under hyperosmotic stress were analyzed. 7E was topically applied to eyes to evaluate the therapeutic effects in the DED animal models. Results IL-20 was significantly upregulated in the tears of patients with DED and in the tears and corneas of DED animal models. Under hyperosmotic stress, IL-20 expression was induced via NFAT5 activation in corneal epithelial cells. 7E suppressed hyperosmotic stress-induced activation of macrophages. IL-20 induced cell death in corneal epithelial cells and 7E protected cells from hyperosmotic stress-induced cell death. Blocking IL-20 signaling with 7E protected mice from BAC-induced, LGE-induced, and DS-induced DED by reducing DED symptoms and inhibiting inflammatory responses, macrophage infiltration, apoptosis, and Th17 populations in the conjunctiva and draining lymph nodes. Conclusions Our results demonstrated the functions of IL-20 in DED and presented a potential therapeutic option for this condition. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12929-022-00821-2.
Collapse
Affiliation(s)
- Hsiao-Hsuan Wang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Wei-Yu Chen
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Hsun Huang
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Ophthalmology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Sheng-Min Hsu
- Department of Ophthalmology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yeou-Ping Tsao
- Department of Medical Research, Mackay Memorial Hospital, Taipei, Taiwan.,Department of Ophthalmology, Mackay Memorial Hospital, Taipei, Taiwan
| | - Yu-Hsiang Hsu
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Research Center of Clinical Medicine, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Ming-Shi Chang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan. .,Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
12
|
Li S, Lu Z, Huang Y, Wang Y, Jin Q, Shentu X, Ye J, Ji J, Yao K, Han H. Anti-Oxidative and Anti-Inflammatory Micelles: Break the Dry Eye Vicious Cycle. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2200435. [PMID: 35435328 PMCID: PMC9189644 DOI: 10.1002/advs.202200435] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 03/13/2022] [Indexed: 05/15/2023]
Abstract
Dry eye disease (DED) impacts ≈30% of the world's population and causes serious ocular discomfort and even visual impairment. Inflammation is one core cause of the DED vicious cycle, a multifactorial deterioration in DED process. However, there are also reactive oxygen species (ROS) regulating inflammation and other points in the cycle from the upstream, leading to treatment failure of current therapies merely targeting inflammation. Accordingly, the authors develop micelle-based eye drops (more specifically p38 mitogen-activated protein kinases (MAPK) inhibitor Losmapimod (Los)-loaded and ROS scavenger Tempo (Tem)-conjugated cationic polypeptide micelles, designated as MTem/Los) for safe and efficient DED management. Cationic MTem/Los improve ocular retention of conjugated water-soluble Tem and loaded water-insoluble Los via electrostatic interaction with negatively charged mucin on the cornea, enabling an increase in therapeutic efficiency and a decrease in dosing frequency. Mechanistically, MTem/Los effectively decrease ROS over-production, reduce the expression of proinflammatory cytokines and chemokines, restrain macrophage proinflammatory phenotypic transformation, and inhibit cell apoptosis. Therapeutically, the dual-functional MTem/Los suppress the inflammatory response, reverse corneal epithelial defect, save goblet cell dysfunction, and recover tear secretion, thus breaking the vicious cycle and alleviating the DED. Moreover, MTem/Los exhibit excellent biocompatibility and tolerability for potential application as a simple and rapid treatment of oxidative stress- and inflammation-induced disorders, including DED.
Collapse
Affiliation(s)
- Su Li
- Eye CenterThe Second Affiliated HospitalSchool of MedicineZhejiang University88 Jiefang RoadHangzhou310009P. R. China
| | - Zhouyu Lu
- Eye CenterThe Second Affiliated HospitalSchool of MedicineZhejiang University88 Jiefang RoadHangzhou310009P. R. China
| | - Yue Huang
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of EducationDepartment of Polymer Science and EngineeringZhejiang UniversityHangzhou310027P. R. China
| | - Yin Wang
- School of PharmacyShanghai Jiao Tong University800 Dongchuan RoadShanghai200240P. R. China
| | - Qiao Jin
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of EducationDepartment of Polymer Science and EngineeringZhejiang UniversityHangzhou310027P. R. China
| | - Xingchao Shentu
- Eye CenterThe Second Affiliated HospitalSchool of MedicineZhejiang University88 Jiefang RoadHangzhou310009P. R. China
- Zhejiang Provincial Key Lab of OphthalmologyZhejiang University88 Jiefang RoadHangzhou310009P. R. China
| | - Juan Ye
- Eye CenterThe Second Affiliated HospitalSchool of MedicineZhejiang University88 Jiefang RoadHangzhou310009P. R. China
- Zhejiang Provincial Key Lab of OphthalmologyZhejiang University88 Jiefang RoadHangzhou310009P. R. China
| | - Jian Ji
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of EducationDepartment of Polymer Science and EngineeringZhejiang UniversityHangzhou310027P. R. China
| | - Ke Yao
- Eye CenterThe Second Affiliated HospitalSchool of MedicineZhejiang University88 Jiefang RoadHangzhou310009P. R. China
- Zhejiang Provincial Key Lab of OphthalmologyZhejiang University88 Jiefang RoadHangzhou310009P. R. China
| | - Haijie Han
- Eye CenterThe Second Affiliated HospitalSchool of MedicineZhejiang University88 Jiefang RoadHangzhou310009P. R. China
- Zhejiang Provincial Key Lab of OphthalmologyZhejiang University88 Jiefang RoadHangzhou310009P. R. China
| |
Collapse
|
13
|
Barros JFF, Sant’Ana AMS, Dias LC, Murashima ADAB, da Silva LECM, Fantucci MZ, Rocha EM. Comparison of the effects of corneal and lacrimal gland denervation on the lacrimal functional unit of rats. Arq Bras Oftalmol 2022; 85:59-67. [PMID: 34586229 PMCID: PMC11826637 DOI: 10.5935/0004-2749.20220008] [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: 02/28/2020] [Accepted: 08/18/2020] [Indexed: 11/20/2022] Open
Abstract
PURPOSE This study aimed to compare the changes in the lacrimal functional unit in the following two models of neurogenic dry eye syndrome: sensory denervation of the cornea versus autonomic denervation of the lacrimal gland. METHODS The neural network supports the lacrimal functional unit. It can be divided into afferent (sensory) and efferent (autonomic) pathways and is affected by severe diseases that compromise the lacrimal functional unit. Male Wistar, 8-week-old rats were divided into the following three groups: 1) control naïve (n=16 animals); 2) autonomic denervation: where rats were subjected to right lacrimal gland nerve ablation and evaluated after 1 and 2 months (1M and 2M) after the procedure (n=7 animals per subgroup, autonomic denervation 1M and autonomic denervation 2M, respectively); 3) sensory denervation induced by 0.2% benzalkonium chloride eye drops, twice a day for 7 days in the right eye (n=10 animals). The corneal sensitivity was measured using the eye wipe test with capsaicin (10 µM). The quantitative real-time PCR was performed to compare the mRNA expressions of proinflammatory cytokines, such as Il-1β, Il-6, Tnf, Mmp9, in the cornea, trigeminal ganglion, and lacrimal gland. In addition, the mRNA of the promitotic factors in the lacrimal gland, such as Bmp7, Runx1, Runx3, Fgf10, and Smad1, was compared. RESULTS Sensory denervation induced corneal hyperalgesia (p=0.001). Sensory denervation and autonomic denervation increased the mRNA of proinflammatory cytokines in the cornea and lacrimal gland (p<0.05), but only sensory denervation increased the mRNA levels of Il-1β and Tnf in the trigeminal ganglion (p<0.05) compared with the control naïve. CONCLUSIONS Autonomic denervation and sensory denervation models can have common features, such as inflammation of different parts of the lacrimal functional unit. However, hyperesthesia and inflammatory markers in the trigeminal ganglion because of sensory denervation and the expression of regenerative mediators in the lacrimal gland owing to autonomic denervation are the distinguishing features of these diseases that can be explored in future studies assessing dry eye syndrome secondary to neural damage of the lacrimal functional unit.
Collapse
Affiliation(s)
- Jacqueline Ferreira Faustino Barros
- Department of Ophthalmology, Otorhinolaryngology, and Head and Neck
Surgery, Faculdade de Medicina de Ribeirão Preto, Universidade de São
Paulo, Ribeirão Preto, SP Brazil
| | - Ariane Mirela Saranzo Sant’Ana
- Department of Ophthalmology, Otorhinolaryngology, and Head and Neck
Surgery, Faculdade de Medicina de Ribeirão Preto, Universidade de São
Paulo, Ribeirão Preto, SP Brazil
| | - Lara Cristina Dias
- Department of Ophthalmology, Otorhinolaryngology, and Head and Neck
Surgery, Faculdade de Medicina de Ribeirão Preto, Universidade de São
Paulo, Ribeirão Preto, SP Brazil
| | - Adriana de Andrade Batista Murashima
- Department of Ophthalmology, Otorhinolaryngology, and Head and Neck
Surgery, Faculdade de Medicina de Ribeirão Preto, Universidade de São
Paulo, Ribeirão Preto, SP Brazil
| | - Lilian Eslaine Costa Mendes da Silva
- Department of Ophthalmology, Otorhinolaryngology, and Head and Neck
Surgery, Faculdade de Medicina de Ribeirão Preto, Universidade de São
Paulo, Ribeirão Preto, SP Brazil
| | - Marina Zilio Fantucci
- Department of Ophthalmology, Otorhinolaryngology, and Head and Neck
Surgery, Faculdade de Medicina de Ribeirão Preto, Universidade de São
Paulo, Ribeirão Preto, SP Brazil
| | - Eduardo Melani Rocha
- Department of Ophthalmology, Otorhinolaryngology, and Head and Neck
Surgery, Faculdade de Medicina de Ribeirão Preto, Universidade de São
Paulo, Ribeirão Preto, SP Brazil
| |
Collapse
|
14
|
Cellulosic Polymers for Enhancing Drug Bioavailability in Ocular Drug Delivery Systems. Pharmaceuticals (Basel) 2021; 14:ph14111201. [PMID: 34832983 PMCID: PMC8621906 DOI: 10.3390/ph14111201] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/15/2021] [Accepted: 11/16/2021] [Indexed: 12/24/2022] Open
Abstract
One of the major impediments to drug development is low aqueous solubility and thus poor bioavailability, which leads to insufficient clinical utility. Around 70–80% of drugs in the discovery pipeline are suffering from poor aqueous solubility and poor bioavailability, which is a major challenge when one has to develop an ocular drug delivery system. The outer lipid layer, pre-corneal, dynamic, and static ocular barriers limit drug availability to the targeted ocular tissues. Biopharmaceutical Classification System (BCS) class II drugs with adequate permeability and limited or no aqueous solubility have been extensively studied for various polymer-based solubility enhancement approaches. The hydrophilic nature of cellulosic polymers and their tunable properties make them the polymers of choice in various solubility-enhancement techniques. This review focuses on various cellulose derivatives, specifically, their role, current status and novel modified cellulosic polymers for enhancing the bioavailability of BCS class II drugs in ocular drug delivery systems.
Collapse
|
15
|
Joo JH, Kim H, Shin JH, Moon SW. Aqueous humor cytokine levels through microarray analysis and a sub-analysis based on optical coherence tomography in wet age-related macular degeneration patients. BMC Ophthalmol 2021; 21:399. [PMID: 34794403 PMCID: PMC8603589 DOI: 10.1186/s12886-021-02152-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 10/18/2021] [Indexed: 11/26/2022] Open
Abstract
Background To identify disease-specific cytokine and growth factor profile differences in the aqueous humor between wet age-related macular degeneration (AMD) patients and age-matched controls and to correlate their levels with the optical coherence tomography (OCT) findings. Methods Aqueous humors were obtained from 13 wet AMD eyes and 10 control eyes. Twenty cytokines and growth factors were measured using a RayBio antibody microarray technology in wet AMD and control eyes. Results The samples obtained from wet AMD patients exhibited a significantly increased expression of MCP-1, MIP-1α, MIP-1β, and vascular endothelial growth factor (VEGF). Subretinal fluid (SRF) patients showed significantly lower levels of proinflammatory cytokines, such as IL-1α and GM-CSF, than those without SRF. Pigment epithelial detachments (PED) patients showed lower levels of inflammatory cytokines, such as GM-CSF, IFN-γ, and TNF-α, than those without PED. Subretinal tissue (SRT) patients showed a higher level of IFN-γ than those without SRT. Compared with the controls, type 1 macular neovascularization (MNV) patients showed increased levels of MCP-1, MIP-1α, and MIP-1β, but not VEGF (p = 0.083). However, type 2 MNV patients showed increased levels of MCP-1 and VEGF (p = 0.040 and p = 0.040). Conclusion Inflammatory cytokines varied according to the type of AMD- and OCT-based parameters. Our observation of low levels of VEGF in patients with type 1 MNV implies that the inhibition of VEGF alone appears to be insufficient treatment for these patients and that cytokines such as MCP-1, MIP-1α, and MIP-1β should be modulated. And the presence of SRF in MNV may be associated with a positive prognosis because we found relatively low levels of proinflammatory cytokines. Supplementary Information The online version contains supplementary material available at 10.1186/s12886-021-02152-6.
Collapse
Affiliation(s)
- Jin-Ho Joo
- Department of Ophthalmology, Kyung Hee University Hospital at Gangdong, 892 Dongnam-ro, Gangdong-gu, Seoul, Republic of Korea
| | - Hyejee Kim
- Barunbit EYE Clinic, Seoul, Republic of Korea
| | - Jae-Ho Shin
- Department of Ophthalmology, Kyung Hee University Hospital at Gangdong, 892 Dongnam-ro, Gangdong-gu, Seoul, Republic of Korea
| | - Sang Woong Moon
- Department of Ophthalmology, Kyung Hee University Hospital at Gangdong, 892 Dongnam-ro, Gangdong-gu, Seoul, Republic of Korea.
| |
Collapse
|
16
|
Lin TL, Cheng C, Zeng WT, Duan F, Pei YH, Liu XP, Shang F, Wu KL. Anti-viral activity of Staphylococcus aureus lysates against herpes simplex virus type-I infection: an in vitro and in vivo study. Int J Ophthalmol 2021; 14:1463-1472. [PMID: 34667721 DOI: 10.18240/ijo.2021.10.01] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 02/05/2021] [Indexed: 02/08/2023] Open
Abstract
AIM To investigate the effect of Staphylococcus aureus (S. aures) lysates (SALs) on herpes simplex virus type-I (HSV1) infection in human corneal epithelial (HCE) cells and in a mouse model of HSV1 keratitis. METHODS HCE, Vero, HeLa, and BV2 cells were infected with HSV1 [HSV1 f strain, HSV1f; HSV-1-H129 with green fluorescent protein (GFP) knock-in, HSV1g]. Pre- or post-infection, SAL at various concentrations was added to the culture medium for 24h. GFP fluorescence in HSV1g or plaque formation by HSV1f were examined. The effects of heat-treated SAL, precooled acetone-precipitated SAL, and SAL subjected to ultrafiltration (100 kDa) were evaluated. The effects of other bacterial components and lysates on HSV1 infection were also tested, including lipoteichoic acid (LTA), peptidoglycan (PGN), staphylococcal protein A (SPA), and α-hemolysin from S. aureus (α-toxin) as well as lysates from a wild-type S. aureus strain, S. epidermidis, and Escherichia coli (W-SAL, SEL, and ECL, respectively). In addition, SAL eye drops were applied topically to BALB/c mice with HSV1 keratitis, followed by in vivo observations. RESULTS The cytopathic effect, plaque formation (HSV1f), and GFP expression (HSV1g) in infected cells were inhibited by SAL in a dose-dependent manner. The active component of SAL (≥100 kDa) was heat-sensitive and retained activity after acetone precipitation. In HSV1g-infected cells, treatment with LTA-sa, α-toxin, PGN-sa, or SPA did not inhibit GFP expression. SAL, W-SAL, and SEL (but not ECL) decreased GFP expression. In mice with HSV1 keratitis, SAL reduced corneal lesions by 71%. CONCLUSION The results of this study demonstrate that SAL can be used to inhibit HSV1 infection, particularly keratitis. Further studies are needed to determine the active components and mechanism underlying the effects of SAL.
Collapse
Affiliation(s)
- Tian-Lan Lin
- Zhongshan Ophthalmic Center, State Key Laboratory of Ophthalmology, Sun Yat-sen University, Guangzhou 510060, Guangdong Province, China
| | - Chao Cheng
- Zhongshan Ophthalmic Center, State Key Laboratory of Ophthalmology, Sun Yat-sen University, Guangzhou 510060, Guangdong Province, China
| | - Wei-Ting Zeng
- Zhongshan Ophthalmic Center, State Key Laboratory of Ophthalmology, Sun Yat-sen University, Guangzhou 510060, Guangdong Province, China
| | - Fang Duan
- Zhongshan Ophthalmic Center, State Key Laboratory of Ophthalmology, Sun Yat-sen University, Guangzhou 510060, Guangdong Province, China
| | - Yin-Hui Pei
- Zhongshan Ophthalmic Center, State Key Laboratory of Ophthalmology, Sun Yat-sen University, Guangzhou 510060, Guangdong Province, China
| | - Xiu-Ping Liu
- Zhongshan Ophthalmic Center, State Key Laboratory of Ophthalmology, Sun Yat-sen University, Guangzhou 510060, Guangdong Province, China
| | - Fu Shang
- Zhongshan Ophthalmic Center, State Key Laboratory of Ophthalmology, Sun Yat-sen University, Guangzhou 510060, Guangdong Province, China
| | - Kai-Li Wu
- Zhongshan Ophthalmic Center, State Key Laboratory of Ophthalmology, Sun Yat-sen University, Guangzhou 510060, Guangdong Province, China
| |
Collapse
|
17
|
Wu JH, Wang TH, Huang JY, Su CC. Ocular Surface Disease in Glaucoma Patients Randomized to Benzalkonium Chloride-Containing Latanoprost and Preservative-Free Bimatoprost. J Ocul Pharmacol Ther 2021; 37:556-564. [PMID: 34610257 DOI: 10.1089/jop.2021.0071] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Purpose: To investigate the influence of benzalkonium chloride (BAK) on ocular surface disease (OSD) in glaucoma patients receiving ocular-hypotensive agent. Methods: Patients were randomized to receive BAK-containing latanoprost (Xalatan) or preservative-free bimatoprost (Lumigan PF). Intraocular pressure (IOP), basal Schirmer's test, noninvasive keratograph tear-breakup time (TBUT), conjunctival redness score (R score), OSD index (OSDI), and corneal Oxford staining were recorded and compared between the 2 groups at 1-month and 4-month visits. The influence of BAK was analyzed by a generalized estimating equation model. Results: We enrolled 74 and 76 eyes treated with latanoprost and bimatoprost, respectively. The IOP decreased in both groups, although greater reduction was observed for latanoprost (13.95 vs. 15.42 mmHg, P = 0.0264). There was a significantly negative association between tear flow and latanoprost use (β = -0.763, P = 0.0243). The first and average TBUT did not show intergroup differences, but the area with unstable tear film increased with latanoprost use and showed marginal significance at 4-month visit (9.33% vs. 5.94% P = 0.055). In both groups, OSDI decreased, whereas Oxford stain increased over time, and R scores showed improvement after transient increase in the first month. The bimatoprost group had significantly worse conjunctival hyperemia, whereas a negative association with conjunctival hyperemia was revealed for latanoprost use (R score-bulbar nasal: β = -0.045, P = 0.0423). Conclusions: BAK-containing latanoprost was associated with decreased tear secretion and may be associated with tear-film instability, whereas bimatoprost was associated with worse conjunctival hyperemia. Ocular surface side effects should be considered when prescribing BAK-containing medication to glaucoma patients.
Collapse
Affiliation(s)
- Jo-Hsuan Wu
- Shiley Eye Institute and Viterbi Family Department of Ophthalmology, University of California, San Diego, California, USA
| | - Tsing-Hong Wang
- Department of Ophthalmology, National Taiwan University Hospital, Taipei, Taiwan.,College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Jehn-Yu Huang
- Department of Ophthalmology, National Taiwan University Hospital, Taipei, Taiwan.,College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chien-Chia Su
- Department of Ophthalmology, National Taiwan University Hospital, Taipei, Taiwan.,College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
18
|
Yamashiro C, Tokuda K, Kobayashi Y, Higashijima F, Yoshimoto T, Ota M, Ogata T, Ashimori A, Kobayashi M, Hatano M, Uchi SH, Wakuta M, Teranishi S, Kimura K. Benzalkonium chloride-induced myofibroblastic transdifferentiation of Tenon's capsule fibroblasts is inhibited by coculture with corneal epithelial cells or by interleukin-10. Sci Rep 2021; 11:16096. [PMID: 34373467 PMCID: PMC8352883 DOI: 10.1038/s41598-021-94852-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 06/04/2021] [Indexed: 11/09/2022] Open
Abstract
Benzalkonium chloride (BAC) is used as a preservative in eyedrops but induces subconjunctival fibrosis that can result in failure of glaucoma surgery. Tenon's capsule fibroblasts in subconjunctival tissue interact with the corneal epithelium through tear fluid. With the use of a coculture system, we have now investigated the effect of human corneal epithelial (HCE) cells on myofibroblastic transdifferentiation of human Tenon fibroblasts (HTFs) induced by BAC (5 × 10-6%). Immunofluorescence and immunoblot analyses revealed that the BAC-induced expression of α smooth muscle actin (αSMA) in HTFs was suppressed by coculture of these cells with HCE cells (p < 0.01). The concentration of interleukin-10 (IL-10) in culture supernatants of BAC-treated HTFs was increased by coculture with HCE cells (17.26-fold, vs. coculure, p < 0.001). Immunofluorescence and immunoblot analyses also showed that exogenous IL-10 (300 pg/ml) suppressed the BAC-induced expression of αSMA by 43.65% (p < 0.05) as well as the nuclear translocation of myocardin-related transcription factor-A (MRTF-A) by 39.32% (p < 0.01) in HTFs cultured alone. Our findings suggest that corneal epithelial cells may protect against subconjunctival fibrosis by maintaining IL-10 levels and preventing the MRTF-A-dependent transdifferentiation of HTFs into myofibroblasts.
Collapse
Affiliation(s)
- Chiemi Yamashiro
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, 755-8505, Japan
| | - Kazuhiro Tokuda
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, 755-8505, Japan
| | - Yuka Kobayashi
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, 755-8505, Japan
| | - Fumiaki Higashijima
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, 755-8505, Japan
| | - Takuya Yoshimoto
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, 755-8505, Japan
| | - Manami Ota
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, 755-8505, Japan
| | - Tadahiko Ogata
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, 755-8505, Japan
| | - Atsushige Ashimori
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, 755-8505, Japan
| | - Masaaki Kobayashi
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, 755-8505, Japan
| | - Makoto Hatano
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, 755-8505, Japan
| | - Sho-Hei Uchi
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, 755-8505, Japan
| | - Makiko Wakuta
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, 755-8505, Japan
| | - Shinichiro Teranishi
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, 755-8505, Japan
| | - Kazuhiro Kimura
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, 755-8505, Japan.
| |
Collapse
|
19
|
Ouyang W, Wu Y, Lin X, Wang S, Yang Y, Tang L, Liu Z, Wu J, Huang C, Zhou Y, Zhang X, Hu J, Liu Z. Role of CD4+ T Helper Cells in the Development of BAC-Induced Dry Eye Syndrome in Mice. Invest Ophthalmol Vis Sci 2021; 62:25. [PMID: 33492357 PMCID: PMC7838551 DOI: 10.1167/iovs.62.1.25] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Purpose To evaluate the role of CD4+ T helper cells in benzalkonium chloride (BAC)-induced ocular surface disorder in C57BL/6 mice. Methods Topical 0.075% BAC was applied twice daily in C57BL/6 mice for 7 consecutive days; PBS-treated and untreated mice served as controls. Adoptive transfer of CD4+ T cells isolated from the BAC-treated mice or PBS-treated mice into nude mice was conducted to identify the roles of CD4+ T cells, with untreated nude mice as controls. Oregon green dextran staining, PAS staining, and the phenol red cotton test were carried out in these two models. The gene and protein levels of T-bet, IFN-γ, RORγt, and IL-17 were detected by quantitative RT-PCR and ELISA, respectively. The activation and subsets of CD4+ T cells were identified by double immunofluorescent staining and flow cytometry. Results An increase in CD4+CD69+, CD4+IFN-γ+, and CD4+IL-17+ cells was induced by BAC in C57BL/6 mice. IFN-γ, IL-17, Th1, Th17, and the transcription factors T-bet and RORγt were increased in BAC-treated mice compared with control mice. In addition, ocular surface damage, including corneal barrier dysfunction, goblet cell loss, and decreased tear production, was induced by BAC. Interestingly, adoptive transfer of CD4+ T cells isolated from BAC-treated mice into nude mice resulted in ocular surface manifestations similar to those of direct topical BAC treatment of C57BL/6 mice, including increased CD4+ T cells, IFN-γ, IL-17, and ocular surface disorders. Conclusions Topical application of BAC induced a dry-eye-like ocular surface disorder partly through the CD4+ T cell-mediated inflammatory response.
Collapse
Affiliation(s)
- Weijie Ouyang
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Yang Wu
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China.,Department of Ophthalmology, Xiamen Branch, Zhongshan Hospital, Fudan University, Xiamen, China
| | - Xiang Lin
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Shoubi Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Yiran Yang
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Liying Tang
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Zhaolin Liu
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Jieli Wu
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Caihong Huang
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Yueping Zhou
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Xiaobo Zhang
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Jiaoyue Hu
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China.,Xiamen University Affiliated Xiamen Eye Center, Xiamen, Fujian, China
| | - Zuguo Liu
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China.,Xiamen University Affiliated Xiamen Eye Center, Xiamen, Fujian, China
| |
Collapse
|
20
|
Yang FM, Fan D, Yang XQ, Zhu FH, Shao MJ, Li Q, Liu YT, Lin ZM, Cao SQ, Tang W, He SJ, Zuo JP. The artemisinin analog SM934 alleviates dry eye disease in rodent models by regulating TLR4/NF-κB/NLRP3 signaling. Acta Pharmacol Sin 2021; 42:593-603. [PMID: 32747720 PMCID: PMC8114933 DOI: 10.1038/s41401-020-0484-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 07/15/2020] [Indexed: 01/22/2023] Open
Abstract
Dry eye disease (DED) is a multifactorial disorder of the tears and ocular surface characterized by manifestations of dryness and irritation. Although the pathogenesis is not fully illuminated, it is recognized that inflammation has a prominent role in the development and deterioration of DED. β-aminoarteether maleate (SM934) is a water-soluble artemisinin derivative with anti-inflammatory and immunosuppressive activities. In this study, we established scopolamine hydrobromide (SCOP)-induced rodent model as well as benzalkonium chloride (BAC)-induced rat model to investigate the therapeutic potential of SM934 for DED. We showed that topical application of SM934 (0.1%, 0.5%) significantly increased tear secretion, maintained the number of conjunctival goblet cells, reduced corneal damage, and decreased the levels of inflammatory mediators (TNF-α, IL-6, IL-10, or IL-1β) in conjunctiva in SCOP-induced and BAC-induced DED models. Moreover, SM934 treatment reduced the accumulation of TLR4-expressing macrophages in conjunctiva, and suppressed the expression of inflammasome components, i.e., myeloid differentiation factor88 (MyD88), Nod-like receptor protein 3 (NLRP3), apoptosis-associated speck-like protein containing CARD (ASC), and cleaved caspase 1. In LPS-treated RAW 264.7 cells, we demonstrated that pretreatment with SM934 (10 μM) impeded the upregulation of TLR4 and downstream NF-κB/NLRP3 signaling proteins. Collectively, artemisinin analog SM934 exerts therapeutic benefits on DED by simultaneously reserving the structural integrity of ocular surface and preventing the corneal and conjunctival inflammation, suggested a further application of SM934 in ophthalmic therapy, especially for DED.
Collapse
Affiliation(s)
- Fang-Ming Yang
- Laboratory of Immunology and Virology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Di Fan
- Laboratory of Immunology and Virology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xiao-Qian Yang
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Feng-Hua Zhu
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Mei-Juan Shao
- Laboratory of Immunology and Virology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Qian Li
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yu-Ting Liu
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ze-Min Lin
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Shi-Qi Cao
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Wei Tang
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shi-Jun He
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Jian-Ping Zuo
- Laboratory of Immunology and Virology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
21
|
Jang J, Kim J, Shin H, Park YG, Joo BJ, Seo H, Won JE, Kim DW, Lee CY, Kim HK, Park JU. Smart contact lens and transparent heat patch for remote monitoring and therapy of chronic ocular surface inflammation using mobiles. SCIENCE ADVANCES 2021; 7:eabf7194. [PMID: 33789904 PMCID: PMC8011975 DOI: 10.1126/sciadv.abf7194] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Accepted: 02/11/2021] [Indexed: 05/24/2023]
Abstract
Wearable electronic devices that can monitor physiological signals of the human body to provide biomedical information have been drawing extensive interests for sustainable personal health management. Here, we report a human pilot trial of a soft, smart contact lens and a skin-attachable therapeutic device for wireless monitoring and therapy of chronic ocular surface inflammation (OSI). As a diagnostic device, this smart contact lens enables real-time measurement of the concentration of matrix metalloproteinase-9, a biomarker for OSI, in tears using a graphene field-effect transistor. As a therapeutic device, we also fabricated a stretchable and transparent heat patch attachable on the human eyelid conformably. Both diagnostic and therapeutic devices can be incorporated using a smartphone for their wireless communications, thereby achieving instantaneous diagnosis of OSI and automated hyperthermia treatments. Furthermore, in vivo tests using live animals and human subjects confirm their good biocompatibility and reliability as a noninvasive, mobile health care solution.
Collapse
Affiliation(s)
- Jiuk Jang
- Nano Science Technology Institute, Department of Materials Science and Engineering, Yonsei University, Seoul 03722, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
| | - Joohee Kim
- Nano Science Technology Institute, Department of Materials Science and Engineering, Yonsei University, Seoul 03722, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
| | - Haein Shin
- Nano Science Technology Institute, Department of Materials Science and Engineering, Yonsei University, Seoul 03722, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
| | - Young-Geun Park
- Nano Science Technology Institute, Department of Materials Science and Engineering, Yonsei University, Seoul 03722, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
- 3KIURI Institute, Yonsei University, Seoul 03722, Republic of Korea
| | - Byung Jun Joo
- Nano Science Technology Institute, Department of Materials Science and Engineering, Yonsei University, Seoul 03722, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
| | - Hunkyu Seo
- Nano Science Technology Institute, Department of Materials Science and Engineering, Yonsei University, Seoul 03722, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
| | - Jong-Eun Won
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
- Department of Dentistry, Korea University Guro Hospital, Seoul 08308, Republic of Korea
- Institute of Clinical Dental Research, Korea University Guro Hospital, Seoul 08308, Republic of Korea
| | - Dai Woo Kim
- Department of Ophthalmology, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu 41944, South Korea
- Bio-Medical Institute, Kyungpook National University Hospital, 130 Dongdeok-ro, Jung-gu, Daegu 41944, South Korea
| | - Chang Young Lee
- School of Energy and Chemical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Hong Kyun Kim
- Department of Ophthalmology, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu 41944, South Korea.
- Bio-Medical Institute, Kyungpook National University Hospital, 130 Dongdeok-ro, Jung-gu, Daegu 41944, South Korea
| | - Jang-Ung Park
- Nano Science Technology Institute, Department of Materials Science and Engineering, Yonsei University, Seoul 03722, Republic of Korea.
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
- 3KIURI Institute, Yonsei University, Seoul 03722, Republic of Korea
| |
Collapse
|
22
|
Wang C, Zhang R, Zhang Q, Jin H, Wei C, Wu C, Mei L, Liu Y, Zhang P. Cytokine Profiles and the Effect of Intravitreal Aflibercept Treatment on Experimental Choroidal Neovascularization. Ophthalmic Res 2020; 65:68-76. [PMID: 33279910 DOI: 10.1159/000513588] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 11/28/2020] [Indexed: 11/19/2022]
Abstract
PURPOSE The purpose of our study was to investigate the profiles of inflammatory cytokines and the macrophage polarization gene in a choroidal neovascularization (CNV) mouse model before and after intravitreal aflibercept treatment. METHODS The CNV mouse model was conducted by laser photocoagulation. A total of 58 cytokines were measured by the multiplex mouse cytokine antibody array. The macrophage polarization genes were tested by reverse transcription polymerase chain reaction. The relationship between the cytokines and the CNV lesion area was analyzed by correlation. RESULTS MIP-1a on day 3 after laser photocoagulation, MCP-5 and Fas-L on day 7, and IL-15 and IL-7 on day 14 were significantly upregulated (p < 0.001, fold change >10.0). After the intravitreal aflibercept treatment, GM-CSF and MCP-1 on day 3 and TIMP-1 on days 7 and 14 were the most significantly upregulated cytokines (p < 0.001, fold change >10.0). MIP-1 on day 3, IL-13 and Fas-L on day 7, and Fas-L on day 14 were the most significantly downregulated cytokines after intravitreal aflibercept treatment (p < 0.001, fold change >5.0). M2 polarization and VEGFA genes were significantly increased in the CNV formation, whereas aflibercept suppressed M2 polarization and VEGFA genes. IL-7 was negatively related to the CNV lesion area on day 14 after intravitreal aflibercept treatment (r = -0.938, p = 0.006). CONCLUSION The inflammatory cytokines and the M1/M2 macrophage genes significantly changed in the CNV mouse model. This result suggests that inflammatory cytokines and macrophages play a critical role in the physiopathology of CNV.
Collapse
Affiliation(s)
- Chen Wang
- Department of Ophthalmology, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China
| | - Rongrong Zhang
- Department of Ophthalmology, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China
| | - Qi Zhang
- Department of Ophthalmology, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China
| | - Huixiang Jin
- Department of Ophthalmology, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China
| | - Chenghua Wei
- Department of Ophthalmology, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China
| | - Changfan Wu
- Department of Ophthalmology, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China
| | - Lixin Mei
- Department of Ophthalmology, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China
| | - Yinping Liu
- Department of Ophthalmology, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China
| | - Pengfei Zhang
- Department of Ophthalmology, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China.,Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institution, Wannan Medical College, Wuhu, China
| |
Collapse
|
23
|
Mohammed I, Kulkarni B, Faraj LA, Abbas A, Dua HS, King AJ. Profiling ocular surface responses to preserved and non-preserved topical glaucoma medications: A 2-year randomized evaluation study. Clin Exp Ophthalmol 2020; 48:973-982. [PMID: 32564453 DOI: 10.1111/ceo.13814] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 06/12/2020] [Accepted: 06/18/2020] [Indexed: 11/27/2022]
Abstract
BACKGROUND Use of topical glaucoma medications has been reported to cause ocular surface (OS) discomfort and inflammation. This study explores the profile of inflammatory cytokines and OS symptoms induced in response to preserved and non-preserved drops. METHODS Prospective, randomized evaluation on 36 treatment-naïve patients over 24 months of three differently preserved glaucoma drop preparations: Preservative-free (PF), polyquad (PQ) and benzalkonium chloride (BAK). Study participants were evaluated at baseline and then at 1, 3, 6, 12 and 24 months while on medication. At each visit, participants completed the OS disease index (OSDI) questionnaire, had basal tear sampling and impression cytology (IC) of the conjunctival epithelium. Quantitative polymerase chain reaction was performed to measure the gene expression of inflammatory cytokines [interleukin (IL)-6, IL-8, IL-10, IL-12A, IL-12B, IL-17A, IL-1β and tumour necrosis factor-α] in the IC samples. Corresponding protein expression of cytokines in tear samples was assessed by the Becton-Dickinson cytometric bead arrays. RESULTS Compared to PF and PQ groups, mRNA and protein expression of IL-6, IL-8 and IL-1β increased in samples from the BAK group in a time-dependent fashion, whereas all other cytokines showed a non-significant increase. In the BAK group, there was a strong correlation between OSDI and the levels of IC/IL-1β (r = .832, R2 = .692 and P = .040); IC/IL-10 (r = .925, R2 = .856 and P = .008) and tear/IL-1β (r = .899, R2 = .808 and P = .014). CONCLUSIONS BAK-preserved topical drops stimulate a sterile inflammatory response on the OS within 3 months which is maintained thereafter, whereas PF-drops and PQ-preserved drops showed no significant OS inflammation.
Collapse
Affiliation(s)
- Imran Mohammed
- Academic Ophthalmology, Division of Clinical Neuroscience, University of Nottingham, Nottingham, UK
| | - Bina Kulkarni
- Academic Ophthalmology, Division of Clinical Neuroscience, University of Nottingham, Nottingham, UK
| | - Lana A Faraj
- Academic Ophthalmology, Division of Clinical Neuroscience, University of Nottingham, Nottingham, UK
| | - Ali Abbas
- Academic Ophthalmology, Division of Clinical Neuroscience, University of Nottingham, Nottingham, UK
| | - Harminder S Dua
- Academic Ophthalmology, Division of Clinical Neuroscience, University of Nottingham, Nottingham, UK
| | - Anthony J King
- Academic Ophthalmology, Division of Clinical Neuroscience, University of Nottingham, Nottingham, UK
| |
Collapse
|
24
|
Choi SW, Cha BG, Kim J. Therapeutic Contact Lens for Scavenging Excessive Reactive Oxygen Species on the Ocular Surface. ACS NANO 2020; 14:2483-2496. [PMID: 31935066 DOI: 10.1021/acsnano.9b10145] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Excessive reactive oxygen species (ROS) play a significant role in the pathogenesis of many eye diseases. Controlling oxidative stress by reducing the amount of ROS is a potential therapeutic strategy for the prevention and treatment of eye diseases, particularly ocular surface diseases. Ceria nanoparticles (CeNPs) have been investigated owing to their efficient ROS-scavenging properties. To overcome the disadvantages of eyedrop administration due to rapid elimination on the surface of the eye and to retain the intrinsic properties of contact lenses, we developed an ROS-scavenging water-soluble CeNP-embedded contact lens (CeNP-CL) for the prevention of ocular surface diseases. The intrinsic ROS-scavenging property of the CeNPs, which mimicked the activities of superoxide dismutase and catalase, was incorporated into polyhydroxyethyl methacrylate-based contact lenses. The CeNP-CL exhibited high transparency and physical properties comparable to those of a commercial contact lens, along with excellent extracellular ROS-scavenging properties. The viabilities of human conjunctival epithelial cells and human meibomian gland epithelial cells were significantly enhanced in the presence of CeNP-CLs, even in media with high H2O2 contents (100 and 500 μM). Additionally, the wearing of CeNP-CLs on the eyes had a protective effect in a mouse model when 3% H2O2 eyedrops were administered. These results indicate the salvaging effect of the CeNP-CL in a high-ROS environment on the ocular surface, which may be helpful for the treatment of ocular surface diseases.
Collapse
Affiliation(s)
- Seung Woo Choi
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology (SAIHST) , Sungkyunkwan University (SKKU) , Suwon 16419 , Republic of Korea
| | - Bong Geun Cha
- School of Chemical Engineering , Sungkyunkwan University (SKKU) , Suwon 16419 , Republic of Korea
| | - Jaeyun Kim
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology (SAIHST) , Sungkyunkwan University (SKKU) , Suwon 16419 , Republic of Korea
- School of Chemical Engineering , Sungkyunkwan University (SKKU) , Suwon 16419 , Republic of Korea
- Biomedical Institute for Convergence at SKKU (BICS) , Sungkyunkwan University (SKKU) , Suwon 16419 , Republic of Korea
- Institute of Quantum Biophysics (IQB) , Sungkyunkwan University (SKKU) , Suwon 16419 , Republic of Korea
| |
Collapse
|
25
|
Kim SJ, Park B, Huh HW, Na YG, Kim M, Han M, Lee H, Pham TMA, Lee HK, Lee JY, Kim CS, Baek JS, Cho CW. Achyranthis radix Extract-Loaded Eye Drop Formulation Development and Novel Evaluation Method for Dry Eye Treatment. Pharmaceutics 2020; 12:pharmaceutics12020165. [PMID: 32079194 PMCID: PMC7076473 DOI: 10.3390/pharmaceutics12020165] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 02/07/2020] [Accepted: 02/13/2020] [Indexed: 12/11/2022] Open
Abstract
Recently, Achyranthis radix extract has been studied as a therapeutic agent for dry eye disease that occurs from fine dust. The aim of this study was the development of Achyranthis radix extract-loaded eye drop formulations using lubricants, generally used for artificial tear eye drops. Ecdysterone was used as a marker compound for Achyranthis radix extract and 1% Achyranthis radix extract solution contained 14.37 ± 0.04 μg/mL of ecdysterone. Before formulation studies, a new method was performed to evaluate pigmentation, which might be caused by eye drops of herbal extract. A comparative study of the water retention ability of each formulation and ability to prevent the death of conjunctival epithelial cells in dry conditions was conducted. Moreover, treatment of Achyranthis radix extract (USL) eye drop formulation exhibited a significant inhibitory effect on inflammation in a concentration-dependent manner. The long-term and accelerated stability tests showed that lubricants could contribute to the stability of herbal extracts in solution. In conclusion, hyaluronic acid showed a good effect on the development of eye drop formulation using Achyranthis radix extracts for treating dry eye disease.
Collapse
Affiliation(s)
- Sung-Jin Kim
- College of Pharmacy and Institute of Drug Research & Development, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Korea; (S.-J.K.); (H.W.H.); (Y.-G.N.); (M.K.); (M.H.); (H.L.); (T.M.A.P.); (H.-K.L.); (J.-Y.L.)
| | - Bongkyun Park
- Clinical Medicine Division, Korea Institute of Oriental Medicine, Daejeon 34054, Korea; (B.P.); (C.-S.K.)
| | - Hyun Wook Huh
- College of Pharmacy and Institute of Drug Research & Development, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Korea; (S.-J.K.); (H.W.H.); (Y.-G.N.); (M.K.); (M.H.); (H.L.); (T.M.A.P.); (H.-K.L.); (J.-Y.L.)
| | - Young-Guk Na
- College of Pharmacy and Institute of Drug Research & Development, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Korea; (S.-J.K.); (H.W.H.); (Y.-G.N.); (M.K.); (M.H.); (H.L.); (T.M.A.P.); (H.-K.L.); (J.-Y.L.)
| | - Minki Kim
- College of Pharmacy and Institute of Drug Research & Development, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Korea; (S.-J.K.); (H.W.H.); (Y.-G.N.); (M.K.); (M.H.); (H.L.); (T.M.A.P.); (H.-K.L.); (J.-Y.L.)
| | - Mingu Han
- College of Pharmacy and Institute of Drug Research & Development, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Korea; (S.-J.K.); (H.W.H.); (Y.-G.N.); (M.K.); (M.H.); (H.L.); (T.M.A.P.); (H.-K.L.); (J.-Y.L.)
| | - Hyunmin Lee
- College of Pharmacy and Institute of Drug Research & Development, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Korea; (S.-J.K.); (H.W.H.); (Y.-G.N.); (M.K.); (M.H.); (H.L.); (T.M.A.P.); (H.-K.L.); (J.-Y.L.)
| | - Thi Mai Anh Pham
- College of Pharmacy and Institute of Drug Research & Development, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Korea; (S.-J.K.); (H.W.H.); (Y.-G.N.); (M.K.); (M.H.); (H.L.); (T.M.A.P.); (H.-K.L.); (J.-Y.L.)
| | - Hong-Ki Lee
- College of Pharmacy and Institute of Drug Research & Development, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Korea; (S.-J.K.); (H.W.H.); (Y.-G.N.); (M.K.); (M.H.); (H.L.); (T.M.A.P.); (H.-K.L.); (J.-Y.L.)
| | - Jae-Young Lee
- College of Pharmacy and Institute of Drug Research & Development, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Korea; (S.-J.K.); (H.W.H.); (Y.-G.N.); (M.K.); (M.H.); (H.L.); (T.M.A.P.); (H.-K.L.); (J.-Y.L.)
| | - Chan-Sik Kim
- Clinical Medicine Division, Korea Institute of Oriental Medicine, Daejeon 34054, Korea; (B.P.); (C.-S.K.)
- Korean Convergence Medicine, University of Science and Technology (UST), Daejeon 34054, Korea
| | - Jong-Suep Baek
- Department of Herbal Medicine Resource, Kangwon National University, 346 Hwangjo-gil, Dogye-eup, Samcheok-si, Gangwon-do 25949, Korea;
| | - Cheong-Weon Cho
- College of Pharmacy and Institute of Drug Research & Development, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Korea; (S.-J.K.); (H.W.H.); (Y.-G.N.); (M.K.); (M.H.); (H.L.); (T.M.A.P.); (H.-K.L.); (J.-Y.L.)
- Correspondence: ; Tel.: +82-42-821-5934
| |
Collapse
|
26
|
Mitochondrial Dysfunctions May Be One of the Major Causative Factors Underlying Detrimental Effects of Benzalkonium Chloride. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:8956504. [PMID: 32104543 PMCID: PMC7035552 DOI: 10.1155/2020/8956504] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 12/23/2019] [Accepted: 01/10/2020] [Indexed: 02/06/2023]
Abstract
Benzalkonium chloride (BAC) is currently the most commonly used antimicrobial preservative in ophthalmic solutions, nasal sprays, and cosmetics. However, a large number of clinical and experimental investigations showed that the topical administration of BAC-containing eye drops could cause a variety of ocular surface changes, from ocular discomfort to potential risk for future glaucoma surgery. BAC-containing albuterol may increase the risk of albuterol-related systemic adverse effects. BAC, commonly present in personal care products, in cosmetic products can induce irritation and dose-dependent changes in the cell morphology. The cationic nature of BAC (it is a quaternary ammonium) suggests that one of the major targets of BAC in the cell may be mitochondria, the only intracellular compartment charged negatively. However, the influence of BAC on mitochondria has not been clearly understood. Here, the effects of BAC on energy parameters of rat liver mitochondria as well as on yeast cells were examined. BAC, being a "weaker" uncoupler, potently inhibited respiration in state 3, diminished the mitochondrial membrane potential, caused opening of the Ca2+/Pi-dependent pore, blocked ATP synthesis, and promoted H2O2 production by mitochondria. BAC triggered oxidative stress and mitochondrial fragmentation in yeast cells. BAC-induced oxidative stress in mitochondria and yeast cells was almost totally prevented by the mitochondria-targeted antioxidant SkQ1; the protective effect of SkQ1 on mitochondrial fragmentation was only partial. Collectively, these data showed that BAC acts adversely on cell bioenergetics (especially on ATP synthesis) and mitochondrial dynamics and that its prooxidant effect can be partially prevented by the mitochondria-targeted antioxidant SkQ1.
Collapse
|
27
|
Qu M, Qi X, Wang Q, Wan L, Li J, Li W, Li Y, Zhou Q. Therapeutic Effects of STAT3 Inhibition on Experimental Murine Dry Eye. Invest Ophthalmol Vis Sci 2020; 60:3776-3785. [PMID: 31503282 DOI: 10.1167/iovs.19-26928] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose To investigate the therapeutic effects of targeting signal transducer and activator of transcription-3 (STAT3) activation on the ocular surface damage of dry eye in mice. Methods Adult Balb/C and C57BL/6 mice with benzalkonium chloride (BAC) treatment, lacrimal gland excision, and meibomian gland dysfunction were used as dry eye models. The levels of phosphorylated STAT3 (p-STAT3) were detected with immunofluorescence staining and Western blotting. STAT3 inhibition was performed by topical application of STAT3 inhibitor S3I-201. Corneal epithelial barrier function, tear production, and conjunctival goblet cell density were quantified with fluorescein sodium staining, phenol red cotton test, and histochemical staining. The expressions of matrix metalloproteinase (MMP)-3/9, TUNEL, and inflammation cytokines were assessed with immunofluorescence staining, qPCR, and ELISA assays. The therapeutic effect of S3I-201 was further compared with the Janus kinase inhibitor tofacitinib and ruxolitinib. Results Elevated levels of nuclear p-STAT3 were detected in the corneal and conjunctival epithelium of three dry eye models. Topical application of S3I-201 improved corneal epithelial barrier function, increased tear production and conjunctival goblet cell density in BAC-induced dry eye mice. Moreover, S3I-201 decreased the expression of MMP-3/9, suppressed the apoptosis of corneal and conjunctival epithelial cells, and reduced the levels of IL-1β, IL-6, IL-17A, and IFN-γ. Compared with tofacitinib and ruxolitinib, the STAT3 inhibitor S3I-201 showed superior improvement of tear production and inflammatory cytokine expression in lacrimal gland. Conclusions Elevated STAT3 activation is involved in the pathogenesis of dry eye, while targeting STAT3 effectively alleviates BAC-induced ocular surface damage.
Collapse
Affiliation(s)
- Mingli Qu
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao, China
| | - Xia Qi
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao, China
| | - Qian Wang
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao, China
| | - Lei Wan
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao, China
| | - Jing Li
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao, China
| | - Wei Li
- Eye Institute of Xiamen University, Xiamen, China
| | - Yingli Li
- Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Qingjun Zhou
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao, China
| |
Collapse
|
28
|
Ocular Surface Changes in Prostaglandin Analogue-Treated Patients. J Ophthalmol 2019; 2019:9798272. [PMID: 31885896 PMCID: PMC6925925 DOI: 10.1155/2019/9798272] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Accepted: 11/25/2019] [Indexed: 11/18/2022] Open
Abstract
Glaucoma is the second leading cause of blindness globally. Reducing intraocular pressure (IOP) has been acknowledged to be the main therapy for glaucoma. Prostaglandin analogues (PGAs) have become the first-line therapy for patients with glaucoma due to their powerful efficacy for lowering (IOP). However, usage of PGAs can also cause several notable side effects, including the changes in ocular surface. The relationship between PGAs and ocular surface changes is complicated and still remains unclear. In the present review, we summarize the recent studies of the effects of PGAs on ocular changes as well as the possible mechanisms that might provide new considerations during clinical medication.
Collapse
|
29
|
Mecum NE, Cyr D, Malon J, Demers D, Cao L, Meng ID. Evaluation of Corneal Damage After Lacrimal Gland Excision in Male and Female Mice. Invest Ophthalmol Vis Sci 2019; 60:3264-3274. [PMID: 31369671 PMCID: PMC6675517 DOI: 10.1167/iovs.18-26457] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose Lacrimal gland excision (LGE) has been utilized in several studies to model aqueous tear deficiency, yet sex as a biological variable has not been factored in to these reports. This study compared corneal pathology in male and female mice following LGE-induced dry eye. Methods An LGE of either the extraorbital lacrimal gland (single LGE) or both the extraorbital and intraorbital lacrimal glands (double LGE) was performed in male and female C57BL/6J and Balb/cJ mice to produce dry eye of graded severity. Following excision, tearing was evaluated with phenol red thread, and corneal fluorescein staining was scored to quantify the severity of damage. Corneas were evaluated for apoptosis by the TUNEL assay and for cell proliferation using Ki67 staining. Furthermore, corneas were harvested and analyzed for macrophages via flow cytometry. Results Baseline tearing levels were similar in male and female mice, and LGE resulted in comparable reductions in tearing with the lowest levels recorded after double LGE. As determined by fluorescein staining, LGE produced more severe damage to the cornea in female C57BL/6J and Balb/cJ mice. Double LGE increased TUNEL and Ki67 staining in the cornea, with greater increases found in female mice. Furthermore, LGE produced a greater increase in the total number of corneal macrophages in female mice. Conclusions These results indicate that female mice are more susceptible to LGE-induced corneal damage. The mechanisms involved in producing these sex differences still need to be elucidated but may involve increased inflammation and macrophage infiltration.
Collapse
Affiliation(s)
- Neal E Mecum
- Center for Excellence in the Neurosciences, University of New England, Biddeford, Maine, United States.,Molecular and Biomedical Sciences, University of Maine, Orono, Maine, United States
| | - Dan Cyr
- Department of Biomedical Sciences, College of Osteopathic Medicine, University of New England, Biddeford, Maine, United States
| | - Jennifer Malon
- Center for Excellence in the Neurosciences, University of New England, Biddeford, Maine, United States
| | - Danielle Demers
- Center for Excellence in the Neurosciences, University of New England, Biddeford, Maine, United States
| | - Ling Cao
- Center for Excellence in the Neurosciences, University of New England, Biddeford, Maine, United States.,Department of Biomedical Sciences, College of Osteopathic Medicine, University of New England, Biddeford, Maine, United States
| | - Ian D Meng
- Center for Excellence in the Neurosciences, University of New England, Biddeford, Maine, United States.,Department of Biomedical Sciences, College of Osteopathic Medicine, University of New England, Biddeford, Maine, United States
| |
Collapse
|
30
|
Zhang R, Park M, Richardson A, Tedla N, Pandzic E, de Paiva CS, Watson S, Wakefield D, Di Girolamo N. Dose-dependent benzalkonium chloride toxicity imparts ocular surface epithelial changes with features of dry eye disease. Ocul Surf 2019; 18:158-169. [PMID: 31740391 DOI: 10.1016/j.jtos.2019.11.006] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 10/15/2019] [Accepted: 11/13/2019] [Indexed: 02/07/2023]
Abstract
PURPOSE Inclusion of the preservative benzalkonium chloride (BAC) in ophthalmic solutions is prevalent, despite the noted potential for exacerbating dry eye disease (DED). Whilst studies incorporating BAC have assessed its' effects as a mouse model of DED, the impact on limbal epithelia is under-studied. Our investigation aimed to comprehensively assess the impact of different BAC dosing regimens and their suitability as a mouse model of DED. METHODS C57BL/6J mice (n = 72) were administered topical BAC (0.05-0.2%) over 7 days. Fluorescein staining, corneal smoothness index, and immuno-histological analyses were applied to determine architectural and cellular changes on the ocular surface following BAC treatment. The effect of BAC (0.0001-0.01%) on cultivated primary mouse corneo-limbal epithelial cells (CLECs) (n = 6) was examined using morphological and functional assays. RESULTS Whilst 0.2% BAC induced severe corneal epithelial defects, 0.1% BAC dispensed once daily over 7 days, induced punctate fluorescein staining without detriment to corneal smoothness. Histochemical staining revealed disorganized basal corneal epithelial cells with enlarged cytoplasmic halos. Furthermore, PAS+ goblet cells were decreased. BAC treatment also modulated K14 expression and distribution within the limbus. In cultured CLEC, BAC triggered cell contraction and vacuolation, increased LDH release and elevated cell necrosis by 4.1-fold. Concentrations of BAC as low as 0.0001% decreased colony formation. CONCLUSIONS This study describes how exposing C57BL/6 mice to BAC induce some clinicopathological features of DED seen in humans, and therefore provides the foundations to explore the consequences on the ocular surface, particularly on limbal epithelia and its' stem cells.
Collapse
Affiliation(s)
- Richard Zhang
- Mechanisms of Disease and Translational Research, School of Medical Sciences, Faculty of Medicine, University of NSW, Sydney, NSW, 2052, Australia
| | - Mijeong Park
- Mechanisms of Disease and Translational Research, School of Medical Sciences, Faculty of Medicine, University of NSW, Sydney, NSW, 2052, Australia
| | - Alexander Richardson
- Mechanisms of Disease and Translational Research, School of Medical Sciences, Faculty of Medicine, University of NSW, Sydney, NSW, 2052, Australia
| | - Nicodemus Tedla
- Mechanisms of Disease and Translational Research, School of Medical Sciences, Faculty of Medicine, University of NSW, Sydney, NSW, 2052, Australia
| | - Elvis Pandzic
- Biomedical Imaging Facility, Mark Wainwright Analytical Centre, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Cintia S de Paiva
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX, 77030, United States
| | - Stephanie Watson
- Save Sight Institute, Discipline of Ophthalmology, Sydney Medical School, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Denis Wakefield
- Mechanisms of Disease and Translational Research, School of Medical Sciences, Faculty of Medicine, University of NSW, Sydney, NSW, 2052, Australia
| | - Nick Di Girolamo
- Mechanisms of Disease and Translational Research, School of Medical Sciences, Faculty of Medicine, University of NSW, Sydney, NSW, 2052, Australia.
| |
Collapse
|
31
|
Effect of a Cationic Surfactant on Microemulsion Globules and Drug Release from Hydrogel Contact Lenses. Pharmaceutics 2019; 11:pharmaceutics11060262. [PMID: 31174291 PMCID: PMC6631260 DOI: 10.3390/pharmaceutics11060262] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 06/01/2019] [Accepted: 06/03/2019] [Indexed: 02/04/2023] Open
Abstract
The present study evaluates the in vitro release of diclofenac sodium (DFNa) from contact lenses based on poly-2-hydroxyethyl methacrylate (pHEMA) hydrogels containing an embedded microemulsion to extend release duration. The oil (ethyl butyrate)-in-water microemulsion systems are prepared with two non-ionic surfactants, Brij 97 or Tween 80, together with a long-alkyl chain cationic surfactant, cetalkonium chloride (CKC). Without CKC, Brij 97 or Tween 80-based microemulsions showed average droplet sizes of 12 nm and 18 nm, respectively. The addition of CKC decreased the average droplet sizes to 2–5 nm for both non-ionic surfactants. Such significant reduction in the average droplet size corresponds to an increase in the DFNa release duration as revealed by the in vitro experiments. Contact lens characterization showed that important properties such as optical transparency and water content of Brij 97-based contact lenses with cationic microemulsions was excellent. However, the optical transparency of the corresponding Tween 80 based contact lenses was unsatisfactory. The results indicate that cationic microemulsion-laden contact lenses can benefit from combinatory effects of microemulsions and cationic surfactant at low CKC weight percentage, e.g., with the release of 70% of the drug in 45, 10, and 7 h for B97-CKC-0.45%, CKC-0.45%, and control lenses, respectively. However, the microemulsion effect on extending DFNa release became negligible at the highest CKC weight percentage (1.8%).
Collapse
|
32
|
Shah M, Cabrera-Ghayouri S, Christie LA, Held KS, Viswanath V. Translational Preclinical Pharmacologic Disease Models for Ophthalmic Drug Development. Pharm Res 2019; 36:58. [PMID: 30805711 PMCID: PMC6394514 DOI: 10.1007/s11095-019-2588-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 02/08/2019] [Indexed: 12/14/2022]
Abstract
Preclinical models of human diseases are critical to our understanding of disease etiology, pathology, and progression and enable the development of effective treatments. An ideal model of human disease should capture anatomical features and pathophysiological mechanisms, mimic the progression pattern, and should be amenable to evaluating translational endpoints and treatment approaches. Preclinical animal models have been developed for a variety of human ophthalmological diseases to mirror disease mechanisms, location of the affected region in the eye and severity. These models offer clues to aid in our fundamental understanding of disease pathogenesis and enable progression of new therapies to clinical development by providing an opportunity to gain proof of concept (POC). Here, we review preclinical animal models associated with development of new therapies for diseases of the ocular surface, glaucoma, presbyopia, and retinal diseases, including diabetic retinopathy and age-related macular degeneration (AMD). We have focused on summarizing the models critical to new drug development and described the translational features of the models that contributed to our understanding of disease pathogenesis and establishment of preclinical POC.
Collapse
Affiliation(s)
- Mihir Shah
- Biological Research, Allergan plc, 2525 Dupont Drive, Irvine, California, 92612, USA
| | - Sara Cabrera-Ghayouri
- Biological Research, Allergan plc, 2525 Dupont Drive, Irvine, California, 92612, USA
| | - Lori-Ann Christie
- Biological Research, Allergan plc, 2525 Dupont Drive, Irvine, California, 92612, USA
| | - Katherine S Held
- Biological Research, Allergan plc, 2525 Dupont Drive, Irvine, California, 92612, USA
| | - Veena Viswanath
- Biological Research, Allergan plc, 2525 Dupont Drive, Irvine, California, 92612, USA.
| |
Collapse
|
33
|
Optimising tear replacement rheology in canine keratoconjunctivitis sicca. Eye (Lond) 2018; 32:195-199. [PMID: 29303147 DOI: 10.1038/eye.2017.272] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 10/08/2017] [Indexed: 12/16/2022] Open
Abstract
Dry eye is a substantial problem in a large number of human and canine patients. Numerous laboratory models for tear deficiency exist using genetically predisposed rodent models, animals treated with topical anti-muscarinics, or those kept in environments with increased air flow to produce the ocular surface changes seen in human patients. Canine keratoconjunctivitis sicca, seen in many thousands of dogs kept as companion animals, can provide a valuable spontaneous model for testing tear replacement medications that might better model disease in human patients, existing as it does in an outbred population that live in the same environments as their owners. Here the development of a crosslinked hyaluronic acid topical drop is described together with the results of trials on dogs with spontaneous keratoconjunctivitis sicca. Although hyaluronic acid in its native form in tear replacement drops shows a Newtonian rheology, the crosslinked product described here behaves in a non-Newtonian manner, with the same shear thinning shown by the tear film itself. The crosslinked product thus shows itself as a potentially valuable tear replacement medication for the human dry eye population as well as for dogs with the same condition.
Collapse
|
34
|
Chen HC, Chen ZY, Wang TJ, Drew VJ, Tseng CL, Fang HW, Lin FH. Herbal Supplement in a Buffer for Dry Eye Syndrome Treatment. Int J Mol Sci 2017; 18:ijms18081697. [PMID: 28771187 PMCID: PMC5578087 DOI: 10.3390/ijms18081697] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Revised: 07/28/2017] [Accepted: 07/29/2017] [Indexed: 12/12/2022] Open
Abstract
Dry eye syndrome (DES) is one of the most common types of ocular diseases. There is a major need to treat DES in a simple yet efficient way. Artificial tears (AT) are the most commonly used agents for treating DES, but are not very effective. Herbal extractions of ferulic acid (FA), an anti-oxidant agent, and kaempferol (KM), an anti-inflammatory reagent, were added to buffer solution (BS) to replace ATs for DES treatment. The cytotoxicity and anti-inflammatory effects were examined in vitro by co-culture with human corneal epithelial cells (HCECs) to obtain the optimal concentration of KM and FA for treating HCECs. Physical properties of BS, such as pH value, osmolality, and refractive index were also examined. Then, rabbits with DES were used for therapeutic evaluation. Tear production, corneal damage, and ocular irritation in rabbits’ eyes were examined. The non-toxic concentrations of KM and FA for HCEC cultivation over 3 days were 1 µM and 100 µM, respectively. Live/dead stain results also show non-toxicity of KM and FA for treating HCECs. Lipopolysaccharide-stimulated HCECs in inflammatory conditions treated with 100 µM FA and 1 µM KM (FA100/KM1) showed lower IL-1B, IL-6, IL-8, and TNFα expression when examined by real-time PCR. The BS with FA100/KM1 had neutral pH, and a similar osmolality and refractive index to human tears. Topical delivery of BS + FA100/KM1 showed no irritation to rabbit eyes. The corneal thickness in the BS + FA100/KM1 treated group was comparable to normal eyes. Results of DES rabbits treated with BS + FA100/KM1 showed less corneal epithelial damage and higher tear volume than the normal group. In conclusion, we showed that the combination of FA (100 µM) and KM (1 µM) towards treating inflamed HCECs had an anti-inflammatory effect, and it is effective in treating DES rabbits when BS is added in combination with these two herbal supplements and used as a topical eye drop.
Collapse
Affiliation(s)
- Hung-Chang Chen
- Department of Chemical Engineering and Biotechnology, National Taipei University of Technology, Taipei 10608, Taiwan.
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan.
| | - Zhi-Yu Chen
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan.
| | - Tsung-Jen Wang
- Department of Ophthalmology, Taipei Medical University Hospital, Taipei 11031, Taiwan.
- Department of Ophthalmology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
| | - Victor J Drew
- International Ph.D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan.
| | - Ching-Li Tseng
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan.
- International Ph.D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan.
| | - Hsu-Wei Fang
- Department of Chemical Engineering and Biotechnology, National Taipei University of Technology, Taipei 10608, Taiwan.
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli 35053, Taiwan.
| | - Feng-Huei Lin
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli 35053, Taiwan.
- Institute of Biomedical Engineering, National Taiwan University, Taipei 10051, Taiwan.
| |
Collapse
|