1
|
Abbasi M, Aghamollaei H, Vaez A, Amani AM, Kamyab H, Chelliapan S, Jamalpour S, Zambrano-Dávila R. Bringing ophthalmology into the scientific world: Novel nanoparticle-based strategies for ocular drug delivery. Ocul Surf 2025; 37:140-172. [PMID: 40147816 DOI: 10.1016/j.jtos.2025.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 03/03/2025] [Accepted: 03/21/2025] [Indexed: 03/29/2025]
Abstract
The distinctive benefits and drawbacks of various drug delivery strategies to supply corneal tissue improvement for sense organs have been the attention of studies worldwide in recent decades. Static and dynamic barriers of ocular tissue prevent foreign chemicals from entering and inhibit the active absorption of therapeutic medicines. The distribution of different medications to ocular tissue is one of the most appealing and demanding tasks for investigators in pharmacology, biomaterials, and ophthalmology, and it is critical for cornea wound healing due to the controlled release rate and increased drug bioavailability. It should be mentioned that the transport of various types of medications into the different sections of the eye, particularly the cornea, is exceedingly challenging because of its distinctive structure and various barriers throughout the eye. Nanoparticles are being studied to improve medicine delivery strategies for ocular disease. Repetitive corneal drug delivery using biodegradable nanocarriers allows a medicine to remain in different parts of the cornea for extended periods of time and thus improve administration route effectiveness. In this review, we discussed eye anatomy, ocular delivery barriers, as well as the emphasis on the biodegradable nanomaterials ranging from organic nanostructures, such as nanomicelles, polymers, liposomes, niosomes, nanowafers, nanoemulsions, nanosuspensions, nanocrystals, cubosomes, olaminosomes, hybridized NPs, dendrimers, bilosomes, solid lipid NPs, nanostructured lipid carriers, and nanofiber to organic nanomaterials like silver, gold, and mesoporous silica nanoparticles. In addition, we describe the nanotechnology-based ophthalmic medications that are presently on the market or in clinical studies. Finally, drawing on current trends and therapeutic approaches, we discuss the challenges that innovative optical drug delivery systems confront and propose future research routes. We hope that this review will serve as a source of motivation and inspiration for developing innovative ophthalmic formulations.
Collapse
Affiliation(s)
- Milad Abbasi
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Hossein Aghamollaei
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Ahmad Vaez
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Mohammad Amani
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hesam Kamyab
- Department of Biomaterials, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, 600077, India; The KU-KIST Graduate School of Energy and Environment, Korea University, 145 Anam-Ro, Seongbuk-Gu, Seoul, 02841, Republic of Korea; Universidad UTE, Quito, 170527, Ecuador.
| | - Shreeshivadasan Chelliapan
- Department of Smart Engineering and Advanced Technology, Faculty of Artificial Intelligence, Universiti Teknologi Malaysia, Jalan Sultan Yahya Petra, 54100, Kuala Lumpur, Malaysia.
| | - Sajad Jamalpour
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, 50603, Malaysia
| | - Renato Zambrano-Dávila
- Universidad UTE, Centro de Investigación en Salud Públicay Epidemiología Clínica (CISPEC), Quito, 170527, Ecuador
| |
Collapse
|
2
|
Lyu D, Ni S, Xu J, Zhu S, Xu JW, Feng Y, Shi C, Xu W. Ocular inoculation of toad venom: toxic cataract and proteomic profiling. Front Med (Lausanne) 2025; 11:1537770. [PMID: 39876868 PMCID: PMC11772289 DOI: 10.3389/fmed.2024.1537770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Accepted: 12/30/2024] [Indexed: 01/31/2025] Open
Abstract
Purpose To report a singular case of cataract caused by toad venom inoculation and to scrutinize the pathological mechanisms through proteomic sequencing of the lens specimen. Methods A young Chinese male presented with progressively deteriorating vision in his right eye subsequent to a history of toad venom inoculation. He was diagnosed with a toxic cataract, and underwent phacoemulsification cataract surgery. Anterior capsule, nucleus, and cortex specimens from the patient (designated as PT_CAP, PT_PHACO, and PT_CTX, respectively) and age-related cataract controls (C_CAP, C_PHACO, and C_CTX, respectively) were collected and subjected to 4D label-free quantitative proteomics. Results A multitude of differentially expressed proteins (DEPs) were identified in the patient's lens compared to those in the controls. Specifically, a total of 204 DEPs were identified in PT_CAP compared to C_CAP, with MYH6, MYL2, MYL3, STAT1, and ANK1 among the foremost regulated DEPs. The DEPs of PT_CAP were principally affiliated with functions including "transportation of small molecules," "regulation of metal ion transport," and "import into cell." A sum of 109 DEPs were delineated in PT_CTX compared to C_CTX, with TPM1 among the top-10 downregulated DEPs. Ninety-five DEPs were pinpointed in PT_PHACO compared to C_PHACO, with hexokinase among the top 10 downregulated DEPs. These proteins were ascertained to be linked with Na+/K+-ATPase activity. Conclusion This study introduced the first documented case of toxic cataract caused by toad venom inoculation. Proteomic sequencing indicated a correlation between cataract and alterations in Na+/K+-ATPase activity, providing insights for the clinical management of ocular toad venom inoculation in subsequent cases.
Collapse
Affiliation(s)
- Danni Lyu
- Eye Center of the 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Zhejiang Provincial Key Lab of Ophthalmology, Hangzhou, Zhejiang, China
| | - Shuang Ni
- Eye Center of the 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Zhejiang Provincial Key Lab of Ophthalmology, Hangzhou, Zhejiang, China
| | - Jia Xu
- Eye Center of the 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Zhejiang Provincial Key Lab of Ophthalmology, Hangzhou, Zhejiang, China
| | - Sha Zhu
- Eye Center of the 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Zhejiang Provincial Key Lab of Ophthalmology, Hangzhou, Zhejiang, China
| | - Jing-Wei Xu
- Eye Center of the 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Zhejiang Provincial Key Lab of Ophthalmology, Hangzhou, Zhejiang, China
| | - Yixuan Feng
- Eye Center of the 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Zhejiang Provincial Key Lab of Ophthalmology, Hangzhou, Zhejiang, China
| | - Ce Shi
- Eye Center of the 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Zhejiang Provincial Key Lab of Ophthalmology, Hangzhou, Zhejiang, China
| | - Wen Xu
- Eye Center of the 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Zhejiang Provincial Key Lab of Ophthalmology, Hangzhou, Zhejiang, China
| |
Collapse
|
3
|
Acott TS, Fautsch MP, Mao W, Ethier CR, Huang AS, Kelley MJ, Aga M, Bhattacharya SK, Borras T, Bovenkamp D, Chowdhury UR, Clark AF, Dibas MI, Du Y, Elliott MH, Faralli JA, Gong H, Herberg S, Johnstone MA, Kaufman PL, Keller KE, Kelly RA, Krizaj D, Kuehn MH, Li HL, Lieberman R, Lin SC, Liu Y, McDonnell FS, McDowell CM, McLellan GJ, Mzyk P, Nair KS, Overby DR, Peters DM, Raghunathan V, Rao PV, Roddy GW, Sharif NA, Shim MS, Sun Y, Thomson BR, Toris CB, Willoughby CE, Zhang HF, Freddo TF, Fuchshofer R, Hill KR, Karimi A, Kizhatil K, Kopcyznski CC, Liton P, Patel G, Peng M, Pattabiraman PP, Prasanna G, Reina-Torres E, Samples EG, Samples JR, Steel CL, Strohmaier CA, Subramanian P, Sugali CK, van Batenburg-Sherwood J, Wong C, Youngblood H, Zode GS, White E, Stamer WD. Consensus Recommendations for Studies of Outflow Facility and Intraocular Pressure Regulation Using Ex Vivo Perfusion Approaches. Invest Ophthalmol Vis Sci 2024; 65:32. [PMID: 39693082 PMCID: PMC11708870 DOI: 10.1167/iovs.65.14.32] [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: 05/04/2024] [Accepted: 09/16/2024] [Indexed: 12/19/2024] Open
Abstract
Intraocular pressure (IOP) elevation is the primary risk factor and currently the main treatable factor for progression of glaucomatous optic neuropathy. In addition to direct clinical and living animal in vivo studies, ex vivo perfusion of anterior segments and whole eyes is a key technique for studying conventional outflow function as it is responsible for IOP regulation. We present well-tested experimental details, protocols, considerations, advantages, and limitations of several ex vivo model systems for studying IOP regulation. These include: (1) perfused whole globes, (2) stationary anterior segment organ culture, (3) perfused human anterior segment organ culture, (4) perfused animal anterior segment organ culture, (5) perfused human corneal rims, and (6) perfused human anterior segment wedges. These methods, with due consideration paid to their strengths and limitations, comprise a set of very strong tools for extending our understanding of IOP regulation.
Collapse
Affiliation(s)
- Ted S. Acott
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - Michael P. Fautsch
- Department of Ophthalmology, Mayo Clinic, Rochester, Minnesota, United States
| | - Weiming Mao
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine, Indianapolis, Indiana, United States
- Departments of Biochemistry and Molecular Biology and Pharmacology and Toxicology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - C. Ross Ethier
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, United States
| | - Alex S. Huang
- Hamilton Glaucoma Center, The Viterbi Family Department of Ophthalmology, Shiley Eye Institute, University of California, San Diego, California, United States
| | - Mary J. Kelley
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - Mini Aga
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - Sanjoy K. Bhattacharya
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami, Florida, United States
| | - Terete Borras
- University of North Carolina, Chapel Hill, North Carolina, United States
| | | | - Uttio Roy Chowdhury
- Department of Ophthalmology, Mayo Clinic, Rochester, Minnesota, United States
| | - Abbot F. Clark
- North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Mohammed I. Dibas
- Glaucoma Research, Ophthalmology Discovery, AbbVie, Irvine, California, United States
| | - Yiqin Du
- Morsani College of Medicine, University of South Florida, Tampa, Florida, United States
| | - Michael H. Elliott
- Department of Ophthalmology and Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Jennifer A. Faralli
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin–Madison, Madison, Wisconsin, United States
| | - Haiyan Gong
- Department of Ophthalmology, Boston University School of Medicine, Boston, Massachusetts, United States
| | - Samuel Herberg
- Department of Ophthalmology & Visual Sciences, SUNY Upstate Medical University, Syracuse, New York, United States
| | - Murray A. Johnstone
- Department of Ophthalmology, University of Washington, Seattle, Washington, United States
| | - Paul L. Kaufman
- Department of Ophthalmology and Visual Sciences, University of Wisconsin–Madison, Madison, Wisconsin, United States
| | - Kate E. Keller
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - Ruth A. Kelly
- Department of Ophthalmology, Duke University, Durham, North Carolina, United States
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, United States
| | - David Krizaj
- Department of Ophthalmology and Visual Sciences, University of Utah, Salt Lake City, Utah, United States
| | - Markus H. Kuehn
- Department Ophthalmology and Visual Sciences, Iowa City VA Center for Prevention and Treatment of Visual Loss, University of Iowa, Iowa City, Iowa, United States
| | - Hoi Lam Li
- Department of Ophthalmology, Boston University School of Medicine, Boston, Massachusetts, United States
| | - Raquel Lieberman
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, Georgia, United States
| | - Shan C. Lin
- Glaucoma Center of San Francisco, San Francisco, California, United States
| | - Yutao Liu
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| | - Fiona S. McDonnell
- John Moran Eye Center, University of Utah, Salt Lake City, Utah, United States
| | - Colleen M. McDowell
- Department of Ophthalmology and Visual Sciences, University of Wisconsin–Madison, Madison, Wisconsin, United States
| | - Gillian J. McLellan
- Department of Ophthalmology and Visual Sciences, University of Wisconsin–Madison, Madison, Wisconsin, United States
- Department of Surgical Sciences, School of Veterinary Medicine, University of Wisconsin–Madison, Madison, Wisconsin, United States
| | - Philip Mzyk
- Department of Ophthalmology, Duke University, Durham, North Carolina, United States
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, United States
| | - Kayarat Saidas Nair
- Deptartment of Ophthalmology, University of California, San Francisco, San Francisco CA, United States
| | - Darryl R. Overby
- Department of Bioengineering, Imperial College of London, London, England
| | - Donna M. Peters
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin–Madison, Madison, Wisconsin, United States
| | - VijayKrishna Raghunathan
- Department of Ophthalmology, Novartis Institutes for BioMedical Research, Cambridge, Massachusetts, United States
| | - Ponugoti Vasantha Rao
- Department of Ophthalmology, Duke University, Durham, North Carolina, United States
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina, United States
| | - Gavin W. Roddy
- Department of Ophthalmology, Mayo Clinic, Rochester, Minnesota, United States
| | - Najam A. Sharif
- Global Alliances and Collaborations, Ophthalmology Innovation Center, Santen Inc., Emeryville, California, United States
| | - Myoung Sup Shim
- Department of Ophthalmology, Duke University, Durham, North Carolina, United States
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, United States
| | - Yang Sun
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, California, United States
| | - Benjamin R. Thomson
- Department of Ophthalmology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States
| | - Carol B. Toris
- Department of Ophthalmology and Visual Sciences, Ohio State University Wexner Medical Center, Columbus, Ohio, United States
| | - Colin E. Willoughby
- Genomic Medicine, Biomedical Sciences Research Institute, Ulster University, Coleraine, Northern Ireland, United Kingdom
| | - Hao F. Zhang
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois, United States
| | - Thomas F. Freddo
- MCP Health Sciences University, Westport, Massachusetts, United States
| | - Rudolf Fuchshofer
- Institute for Human Anatomy and Embryology, University of Regensburg, Regensburg, Germany
| | - Kamisha R. Hill
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, Georgia, United States
| | - Alireza Karimi
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - Krishnakumar Kizhatil
- Department of Ophthalmology and Visual Science, Ohio State University, Columbus, Ohio, United States
| | | | - Paloma Liton
- Department of Ophthalmology, Duke University, Durham, North Carolina, United States
| | - Gaurang Patel
- North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, Texas, United States
- Department of Ophthalmology and Genetics Medicine Research, Regeneron Pharmaceuticals, Inc., Tarrytown, New York, United States
| | - Michael Peng
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine, Indianapolis, Indiana, United States
- Departments of Biochemistry and Molecular Biology and Pharmacology and Toxicology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - Padmanabhan P. Pattabiraman
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - Ganesh Prasanna
- Department of Ophthalmology, Novartis Institutes for BioMedical Research, Cambridge, Massachusetts, United States
| | - Ester Reina-Torres
- Department of Bioengineering, Imperial College of London, London, England
| | | | - John R. Samples
- Washington State College of Medicine, Spokane, Washington, United States
| | | | - Clemens A. Strohmaier
- Department of Ophthalmology and Optometry, Kepler University Hospital, Johannes Kepler University, Linz, Austria
| | | | - Chenna Kesavulu Sugali
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine, Indianapolis, Indiana, United States
- Departments of Biochemistry and Molecular Biology and Pharmacology and Toxicology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | | | - Cydney Wong
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, United States
| | - Hannah Youngblood
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, Georgia, United States
| | - Gulab S. Zode
- Department of Ophthalmology, University of California at Irvine, Irvine, California, United States
| | - Elizabeth White
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - W. Daniel Stamer
- Department of Ophthalmology, Duke University, Durham, North Carolina, United States
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, United States
| |
Collapse
|
4
|
Higashide M, Watanabe M, Sato T, Umetsu A, Nishikiori N, Ogawa T, Furuhashi M, Ohguro H. FABP5 Is a Possible Factor for the Maintenance of Functions of Human Non-Pigmented Ciliary Epithelium Cells. Int J Mol Sci 2024; 25:9285. [PMID: 39273233 PMCID: PMC11394871 DOI: 10.3390/ijms25179285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 08/05/2024] [Accepted: 08/22/2024] [Indexed: 09/15/2024] Open
Abstract
To elucidate the possible biological roles of fatty acid-binding protein 5 (FABP5) in the intraocular environment, the cells from which FABP5 originates were determined by using four different intraocular tissue-derived cell types including human non-pigmented ciliary epithelium (HNPCE) cells, retinoblastoma (RB) cells, adult retinal pigment epithelial19 (ARPE19) cells and human ocular choroidal fibroblast (HOCF) cell lines, and the effects of FABP ligand 6, a specific inhibitor for FABP5 and FABP7 were analyzed by RNA sequencing and seahorse cellular metabolic measurements. Among these four different cell types, qPCR analysis showed that FABP5 was most prominently expressed in HNPCE cells, in which no mRNA expression of FABP7 was detected. In RNA sequencing analysis, 166 markedly up-regulated and 198 markedly down-regulated differentially expressed genes (DEGs) were detected between non-treated cells and cells treated with FABP ligand 6. IPA analysis of these DEGs suggested that FABP5 may be involved in essential roles required for cell development, cell survival and cell homeostasis. In support of this possibility, both mitochondrial and glycolytic functions of HNPCE cells, in which mRNA expression of FABP5, but not that of FABP7, was detected, were shown by using a Seahorse XFe96 Bioanalyzer to be dramatically suppressed by FABP ligand 6-induced inhibition of the activity of FABP5. Furthermore, in IPA upstream analysis, various unfolded protein response (UPR)-related factors were identified as upstream and causal network master regulators. Analysis by qPCR analysis showed significant upregulation of the mRNA expression of most of UPR-related factors and aquaporin1 (AQP1). The findings in this study suggest that HNPCE is one of intraocular cells producing FABP5 and may be involved in the maintenance of UPR and AQP1-related functions of HNPCE.
Collapse
Affiliation(s)
- Megumi Higashide
- Departments of Ophthalmology, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Megumi Watanabe
- Departments of Ophthalmology, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Tatsuya Sato
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
- Department of Cellular Physiology and Signal Transduction, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Araya Umetsu
- Departments of Ophthalmology, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Nami Nishikiori
- Departments of Ophthalmology, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Toshifumi Ogawa
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
- Department of Cellular Physiology and Signal Transduction, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Masato Furuhashi
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Hiroshi Ohguro
- Departments of Ophthalmology, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| |
Collapse
|
5
|
Fang Z, Bi S, Brown JD, Chen J, Pan T. Microfluidics in the eye: a review of glaucoma implants from an engineering perspective. LAB ON A CHIP 2023; 23:4736-4772. [PMID: 37847237 DOI: 10.1039/d3lc00407d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2023]
Abstract
Glaucoma is a progressive optic neuropathy in the eye, which is a leading cause of irreversible blindness worldwide and currently affects over 70 million individuals. Clinically, intraocular pressure (IOP) reduction is the only proven treatment to halt the progression of glaucoma. Microfluidic devices such as glaucoma drainage devices (GDDs) and minimally invasive glaucoma surgery (MIGS) devices are routinely used by ophthalmologists to manage elevated IOP, by creating an artificial pathway for the over-accumulated aqueous humor (AH) in a glaucomatous eye, when the natural pathways are severely blocked. Herein, a detailed modelling and analysis of both the natural microfluidic pathways of the AH in the eye and artificial microfluidic pathways formed additionally by the various glaucoma implants are conducted to provide an insight into the causes of the IOP abnormality and the improvement schemes of current implant designs. The mechanisms of representative glaucoma implants have been critically reviewed from the perspective of microfluidics, and we have categorized the current implants into four groups according to the targeted drainage sites of the AH, namely Schlemm's canal, suprachoroidal space, subconjunctival space, and ocular surface. In addition, we propose to divide the development and evolution of glaucoma implant designs into three technological waves, which include microtube (1st), microvalve (2nd) and microsystem (3rd). With the emerging trends of minimal invasiveness and artificial intelligence in the development of medical implants, we envision that a comprehensive glaucoma treatment microsystem is on the horizon, which is featured with active and wireless control of IOP, real-time continuous monitoring of IOP and aqueous rate, etc. The current review could potentially cast light on the unmatched needs, challenges, and future directions of the microfluidic structural and functional designs of glaucoma implants, which would enable an enhanced safety profile, reduced complications, increased efficacy of lowering IOP and reduced IOP fluctuations, closed-loop and on-demand control of IOP, etc.
Collapse
Affiliation(s)
- Zecong Fang
- Bionic Sensing and Intelligence Center (BSIC), Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China.
| | - Shuzhen Bi
- Center for Intelligent Medical Equipment and Devices (iMED), University of Science and Technology of China, Suzhou, Jiangsu, 215123, China
| | | | - Junyi Chen
- Department of Ophthalmology and Visual Science, Eye and ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, 200031, China
- NHC Key Laboratory of Myopia, Chinese Academy of Medical Sciences, and Shanghai Key Laboratory of Visual Impairment and Restoration (Fudan University), Shanghai, 200031, China
| | - Tingrui Pan
- Bionic Sensing and Intelligence Center (BSIC), Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China.
- Center for Intelligent Medical Equipment and Devices (iMED), University of Science and Technology of China, Suzhou, Jiangsu, 215123, China
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, Anhui, 230026, China
| |
Collapse
|
6
|
Hirono K, Inoue M, Tanaka S, Uchio E, Yanagi Y, Kadonosono K. ANTERIOR SEGMENT OPTICAL COHERENCE TOMOGRAPHY IN DETERMINATION OF ENTRY SITE FOR VITRECTOMY IN HIGHLY MYOPIC EYES. Retina 2023; 43:733-738. [PMID: 36812416 PMCID: PMC10125124 DOI: 10.1097/iae.0000000000003736] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
PURPOSE To evaluate the efficacy of anterior segment optical coherent tomography (AS OCT) in estimating the length of the pars plana and optimizing the sclerotomy entry site in vitrectomy for highly myopic eyes, facilitating membrane peeling. METHODS Twenty-three eyes with myopic traction maculopathy were studied. The pars plana was examined using two methods: preoperative AS OCT and intraoperative measurement. The distance from the limbus to the ora serrata in two groups was measured to compare differences in length. The actual length of the entry site from the limbus and forceps used were noted in all eyes studied. RESULTS The mean axial length was 29.2 ± 2.3 mm for all 23 eyes. The average length between the limbus and ora serrata measured with AS OCT and intraoperative examination was 6,710 µ m (SD ± 459) and 6,671 µ m (SD ± 402), respectively, in the superotemporal region ( P > 0.05), and 6,340 µ m (SD ± 321) and 6,204 µ m (SD ± 402), respectively, in the superonasal region ( P >0.05). The mean length of the entry site from the limbus was 6.2 mm, and 28-mm forceps were used in 17 of 23 eyes (77%). CONCLUSION The length of the pars plana varies depending on the axial length of the eye. Preoperative AS OCT enables accurate measurement of the pars plana in eyes with high myopia. AS OCT examination can help determine the optimal site for sclerotomy, allowing easier access to the macular region for membrane peeling in highly myopic eyes.
Collapse
Affiliation(s)
- Kazushi Hirono
- Department of Ophthalmology and Micro-technology, Yokohama City University; and
| | - Maiko Inoue
- Department of Ophthalmology and Micro-technology, Yokohama City University; and
| | - Shin Tanaka
- Department of Ophthalmology and Micro-technology, Yokohama City University; and
| | | | - Yasuo Yanagi
- Department of Ophthalmology and Micro-technology, Yokohama City University; and
| | - Kazuaki Kadonosono
- Department of Ophthalmology and Micro-technology, Yokohama City University; and
| |
Collapse
|
7
|
Mechanical Stretch Activates TRPV4 and Hemichannel Responses in the Nonpigmented Ciliary Epithelium. Int J Mol Sci 2023; 24:ijms24021673. [PMID: 36675184 PMCID: PMC9865367 DOI: 10.3390/ijms24021673] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/05/2023] [Accepted: 01/12/2023] [Indexed: 01/19/2023] Open
Abstract
Previously, we reported a mechanosensitive ion channel, TRPV4, along with functional connexin hemichannels on the basolateral surface of the ocular nonpigmented ciliary epithelium (NPE). In the lens, TRPV4-mediated hemichannel opening is part of a feedback loop that senses and respond to swelling. The present study was undertaken to test the hypothesis that TRPV4 and hemichannels in the NPE respond to a mechanical stimulus. Porcine NPE cells were cultured on flexible membranes to study effects of cyclic stretch and ATP release was determined by a luciferase assay. The uptake of propidium iodide (PI) was measured as an indicator of hemichannel opening. NPE cells subjected to cyclic stretch for 1-10 min (10%, 0.5 Hz) displayed a significant increase in ATP release into the bathing medium. In studies where PI was added to the bathing medium, the same stretch stimulus increased cell PI uptake. The ATP release and PI uptake responses to stretch both were prevented by a TRPV4 antagonist, HC067047 (10 µM), and a connexin mimetic peptide, Gap 27 (200µm). In the absence of a stretch stimulus, qualitatively similar ATP release and PI uptake responses were observed in cells exposed to the TRPV4 agonist GSK1016790A (10 nM), and Gap 27 prevented the responses. Cells subjected to an osmotic swelling stimulus (hypoosmotic medium: 200 mOsm) also displayed a significant increase in ATP release and PI uptake and the responses were abolished by TRPV4 inhibition. The findings point to TRPV4-dependent connexin hemichannel opening in response to mechanical stimulus. The TRPV4-hemichannel mechanism may act as a mechanosensor that facilitates the release of ATP and possibly other autocrine or paracrine signaling molecules that influence fluid (aqueous humor) secretion by the NPE.
Collapse
|
8
|
Activity-Dependent Neuroprotective Protein (ADNP): An Overview of Its Role in the Eye. Int J Mol Sci 2022; 23:ijms232113654. [PMID: 36362439 PMCID: PMC9658893 DOI: 10.3390/ijms232113654] [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: 10/04/2022] [Revised: 11/02/2022] [Accepted: 11/06/2022] [Indexed: 11/09/2022] Open
Abstract
Vision is one of the dominant senses in humans and eye health is essential to ensure a good quality of life. Therefore, there is an urgent necessity to identify effective therapeutic candidates to reverse the progression of different ocular pathologies. Activity-dependent neuroprotective protein (ADNP) is a protein involved in the physio-pathological processes of the eye. Noteworthy, is the small peptide derived from ADNP, known as NAP, which shows protective, antioxidant, and anti-apoptotic properties. Herein, we review the current state of knowledge concerning the role of ADNP in ocular pathologies, while providing an overview of eye anatomy.
Collapse
|
9
|
Schwarz MM, Connors KA, Davoli KA, McMillen CM, Albe JR, Hoehl RM, Demers MJ, Ganaie SS, Price DA, Leung DW, Amarasinghe GK, McElroy AK, Reed DS, Hartman AL. Rift Valley Fever Virus Infects the Posterior Segment of the Eye and Induces Inflammation in a Rat Model of Ocular Disease. J Virol 2022; 96:e0111222. [PMID: 36194021 PMCID: PMC9599513 DOI: 10.1128/jvi.01112-22] [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: 07/17/2022] [Accepted: 09/16/2022] [Indexed: 01/24/2023] Open
Abstract
People infected with the mosquito-borne Rift Valley fever virus (RVFV) can suffer from eye-related problems resulting in ongoing vision issues or even permanent blindness. Despite ocular disease being the most frequently reported severe outcome, it is vastly understudied compared to other disease outcomes caused by RVFV. Ocular manifestations of RVFV include blurred vision, uveitis, and retinitis. When an infected individual develops macular or paramacular lesions, there is a 50% chance of permanent vision loss in one or both eyes. The cause of blinding ocular pathology remains unknown in part due to the lack of a tractable animal model. Using 3 relevant exposure routes, both subcutaneous (SC) and aerosol inoculation of Sprague Dawley rats led to RVFV infection of the eye. Surprisingly, direct inoculation of the conjunctiva did not result in successful ocular infection. The posterior segment of the eye, including the optic nerve, choroid, ciliary body, and retina, were all positive for RVFV antigen in SC-infected rats, and live virus was isolated from the eyes. Proinflammatory cytokines and increased leukocyte counts were also found in the eyes of infected rats. Additionally, human ocular cell lines were permissive for Lrp1-dependent RVFV infection. This study experimentally defines viral tropism of RVFV in the posterior segment of the rat eye and characterizes virally-mediated ocular inflammation, providing a foundation for evaluation of vaccines and therapeutics to protect against adverse ocular outcomes. IMPORTANCE Rift Valley fever virus (RVFV) infection leads to eye damage in humans in up to 10% of reported cases. Permanent blindness occurs in 50% of individuals with significant retinal scarring. Despite the prevalence and severity of this outcome, very little is known about the mechanisms of pathogenesis. We addressed this gap by developing a rodent model of ocular disease. Subcutaneous infection of Sprague Dawley rats resulted in infection of the uvea, retina, and optic nerve along with the induction of inflammation within the posterior eye. Infection of human ocular cells induced inflammatory responses and required host entry factors for RVFV infection similar to rodents. This work provides evidence of how RVFV infects the eye, and this information can be applied to help mitigate the devastating outcomes of RVF ocular disease through vaccines or treatments.
Collapse
Affiliation(s)
- Madeline M. Schwarz
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Infectious Diseases and Microbiology, School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Kaleigh A. Connors
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Infectious Diseases and Microbiology, School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Katherine A. Davoli
- Ophthalmic and Visual Sciences Research Center, Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Cynthia M. McMillen
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Infectious Diseases and Microbiology, School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Joseph R. Albe
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Ryan M. Hoehl
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Matthew J. Demers
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Safder S. Ganaie
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - David A. Price
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Daisy W. Leung
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
- Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Gaya K. Amarasinghe
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Anita K. McElroy
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Division of Pediatric Infectious Disease, Department of Pediatrics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Douglas S. Reed
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Amy L. Hartman
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Infectious Diseases and Microbiology, School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
10
|
Lou B, Zeng L, Gao X, Qian X, Li JJ, Gu X, Liu Z, Liu K, Chen X, Lin X, Zhang F. A single-cell transcriptomic atlas of the human ciliary body. Cell Mol Life Sci 2022; 79:528. [PMID: 36163311 PMCID: PMC9512889 DOI: 10.1007/s00018-022-04559-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 09/08/2022] [Accepted: 09/12/2022] [Indexed: 12/02/2022]
Abstract
The ciliary body critically contributes to the ocular physiology with multiple responsibilities in the production of aqueous humor, vision accommodation and intraocular immunity. Comparatively little work, however, has revealed the single-cell molecular taxonomy of the human ciliary body required for studying these functionalities. In this study, we report a comprehensive atlas of the cellular and molecular components of human ciliary body as well as their interactions using single-cell RNA sequencing (scRNAseq). Cluster analysis of the transcriptome of 14,563 individual ciliary cells from the eyes of 3 human donors identified 14 distinct cell types, including the ciliary epithelium, smooth muscle, vascular endothelial cell, immune cell and other stromal cell populations. Cell-type discriminative gene markers were also revealed. Unique gene expression patterns essential for ciliary epithelium-mediated aqueous humor inflow and ciliary smooth muscle contractility were identified. Importantly, we discovered the transitional states that probably contribute to the transition of ciliary macrophage into retina microglia and verified no lymphatics in the ciliary body. Moreover, the utilization of CellPhoneDB allowed us to systemically infer cell–cell interactions among diverse ciliary cells including those that potentially participate in the pathogenesis of glaucoma and uveitis. Altogether, these new findings provide insights into the regulation of intraocular pressure, accommodation reflex and immune homeostasis under physiological and pathological conditions.
Collapse
Affiliation(s)
- Bingsheng Lou
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Lei Zeng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Xinbo Gao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Xiaobing Qian
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Jing Jing Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Xinyu Gu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Zheng Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Keli Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Xun Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Xiaofeng Lin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou, 510060, China.
| | - Feng Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou, 510060, China.
| |
Collapse
|
11
|
Dvoriashyna M, Foss AJE, Gaffney EA, Repetto R. A Mathematical Model of Aqueous Humor Production and Composition. Invest Ophthalmol Vis Sci 2022; 63:1. [PMID: 35917134 PMCID: PMC9358295 DOI: 10.1167/iovs.63.9.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose We develop a mathematical model that predicts aqueous humor (AH) production rate by the ciliary processes and aqueous composition in the posterior chamber (PC), with the aim of estimating how the aqueous production rate depends on the controlling parameters and how it can be manipulated. Methods We propose a compartmental mathematical model that considers the stromal region, ciliary epithelium, and PC. All domains contain an aqueous solution with different chemical species. We impose the concentration of all species on the stromal side and exploit the various ion channels present on the cell membrane to compute the water flux produced by osmosis, the solute concentrations in the AH and the transepithelial potential difference. Results With a feasible set of parameters, the model predictions of water flux from the stroma to the PC and of the solute concentrations in the AH are in good agreement with measurements. Key parameters which impact the aqueous production rate are identified. A relevant role is predicted to be played by cell membrane permeability to \(\text{K}^+\) and \(\text{Cl}^-\), by the level of transport due to the Na+-H+ exchanger and to the co-transporter of Na+/K+/2Cl-; and by carbonic anhydrase. Conclusions The mathematical model predicts the formation and composition of AH, based on the structure of the ciliary epithelium. The model provides insight into the physical processes underlying the functioning of drugs that are adopted to regulate the aqueous production. It also suggests ion channels and cell membrane properties that may be targeted to manipulate the aqueous production rate.
Collapse
Affiliation(s)
- Mariia Dvoriashyna
- Department of Applied Mathematics and Theoretical Physics, University of Cambridge, United Kingdom.,Mathematical Institute, University of Oxford, United Kingdom
| | - Alexander J E Foss
- Department of Ophthalmology, Nottingham University Hospitals NHS Trust, United Kingdom
| | | | - Rodolfo Repetto
- Department of Civil, Chemical and Environmental Engineering, University of Genoa, Italy
| |
Collapse
|
12
|
TANABE KAZUHIKO, KIMURA ITARU, OKAMOTO HARU, CHI ZAILONG, AKAHORI MASAKAZU, SHIMOZAWA NOBUHIRO, EBIHARA NOBUYUKI, MURAKAMI AKIRA, IWATA TAKESHI. The Expression of Rab8, Ezrin, Radixin and Moesin in the Ciliary Body of Cynomolgus Monkeys. JUNTENDO IJI ZASSHI = JUNTENDO MEDICAL JOURNAL 2022; 68:339-351. [PMID: 39021423 PMCID: PMC11250011 DOI: 10.14789/jmj.jmj21-0042-oa] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 03/14/2022] [Indexed: 07/20/2024]
Abstract
Purpose The purpose of this study was to determine what proteins are present in the ciliary body (CB). To accomplish this, we conducted a proteomic analysis of the CB of cynomolgus monkeys. We also determined the location of the proteins in CB by immunohistology. Methods The eyes of euthanized cynomolgus monkeys were enucleated, and the CB, were isolated from the eyes. Proteins were extracted from the CB and determined by liquid chromatography-mass spectrometry. Separated CB epithelial cells were cultured, and the proteins expressed in the CB were determined by Western blotting. The location of these proteins in the CB was determined by immunohistochemical staining. We also investigated whether adding dexamethasone to the culture medium changed protein expression by the epithelial cells. Results Proteomic analysis of the CBs showed that 813 proteins were expressed in the epithelium and stroma. These proteins included the small guanosine triphosphate-binding protein Rab8 and the ezrin/radixin/moesin (ERM) family. Tissue and immunohistological staining confirmed the colocalization of these proteins in non-pigmented CB epithelium. Western blotting of cultured CB epithelial cell lysates showed a tendency that adding dexamethasone changed Rab8 protein expression levels. Conclusions Proteomic analysis of CBs identified several proteins involved in the transport and secretion of proteins. These proteins may be involved in the production of aqueous humor and protein secretion by the CB.
Collapse
Affiliation(s)
| | - ITARU KIMURA
- Corresponding author: Itaru Kimura, Division on Molecular & Cellular Biology, National Institute of Sensory Organs, National Hospital Organization Tokyo Medical Center, 2-5-1 Higashigaoka, Meguro-ku, Tokyo 152-8902 Japan, TEL/FAX: +81-3-3411-1026 E-mail:
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Lam MR, Dong P, Shokrollahi Y, Gu L, Suh DW. Finite Element Analysis of Soccer Ball-Related Ocular and Retinal Trauma and Comparison with Abusive Head Trauma. OPHTHALMOLOGY SCIENCE 2022; 2:100129. [PMID: 36249696 PMCID: PMC9560646 DOI: 10.1016/j.xops.2022.100129] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 01/11/2022] [Accepted: 02/14/2022] [Indexed: 11/19/2022]
Abstract
Purpose Design Participants Methods Main Outcome Measures Results Conclusions
Collapse
Affiliation(s)
- Matthew R. Lam
- Creighton University School of Medicine, Omaha, Nebraska
- Correspondence: Matthew R. Lam, Creighton University School of Medicine, 2500 California Plaza, Omaha, NE 68178.
| | - Pengfei Dong
- Florida Institute of Technology, Department of Biomedical and Chemical Engineering & Department of Mechanical Engineering, Melbourne, Florida
| | - Yasin Shokrollahi
- Florida Institute of Technology, Department of Biomedical and Chemical Engineering & Department of Mechanical Engineering, Melbourne, Florida
| | - Linxia Gu
- Florida Institute of Technology, Department of Biomedical and Chemical Engineering & Department of Mechanical Engineering, Melbourne, Florida
| | - Donny W. Suh
- Gavin Herbert Eye Institute, University of California at Irvine, Department of Ophthalmology and Visual Sciences, Irvine, California
| |
Collapse
|
14
|
Fotesko K, Thomsen BSV, Kolko M, Vohra R. Girl Power in Glaucoma: The Role of Estrogen in Primary Open Angle Glaucoma. Cell Mol Neurobiol 2022; 42:41-57. [PMID: 33040237 PMCID: PMC11441221 DOI: 10.1007/s10571-020-00965-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 09/22/2020] [Indexed: 12/24/2022]
Abstract
Estrogen is essential in maintaining various physiological features in women, and a decline in estrogen levels are known to give rise to numerous unfortunate symptoms associated with menopause. To alleviate these symptoms hormone replacement therapy with estrogen is often used, and has been shown to be fruitful in improving quality of life in women suffering from postmenopausal discomforts. An often forgotten condition associated with menopause is the optic nerve disorder, glaucoma. Thus, estrogen may also have an impact in maintaining the retinal ganglion cells (RGCs), which make up the optic nerve, thereby preventing glaucomatous neurodegeneration. This review aims to provide an overview of possible associations of estrogen and the glaucoma subtype, primary open-angle glaucoma (POAG), by evaluating the current literature through a PubMed-based literature search. Multiple in vitro and in vivo studies of RGC protection, as well as clinical and epidemiological data concerning the well-defined retinal neurodegenerative disorder POAG have been reviewed. Over all, deficiencies in retinal estrogen may potentially instigate RGC loss, visual disability, and eventual blindness. Estrogen replacement therapy may therefore be a beneficial future treatment. However, more studies are needed to confirm the relevance of estrogen in glaucoma prevention.
Collapse
Affiliation(s)
- Kyrylo Fotesko
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | | | - Miriam Kolko
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark.
- Department of Ophthalmology, Rigshospitalet-Glostrup, Glostrup, Denmark.
| | - Rupali Vohra
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark.
- Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
15
|
Karamichos D, Escandon P, Vasini B, Nicholas SE, Van L, Dang DH, Cunningham RL, Riaz KM. Anterior pituitary, sex hormones, and keratoconus: Beyond traditional targets. Prog Retin Eye Res 2021; 88:101016. [PMID: 34740824 PMCID: PMC9058044 DOI: 10.1016/j.preteyeres.2021.101016] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 10/15/2021] [Accepted: 10/18/2021] [Indexed: 12/13/2022]
Abstract
"The Diseases of the Horny-coat of The Eye", known today as keratoconus, is a progressive, multifactorial, non-inflammatory ectatic corneal disorder that is characterized by steepening (bulging) and thinning of the cornea, irregular astigmatism, myopia, and scarring that can cause devastating vision loss. The significant socioeconomic impact of the disease is immeasurable, as patients with keratoconus can have difficulties securing certain jobs or even joining the military. Despite the introduction of corneal crosslinking and improvements in scleral contact lens designs, corneal transplants remain the main surgical intervention for treating keratoconus refractory to medical therapy and visual rehabilitation. To-date, the etiology and pathogenesis of keratoconus remains unclear. Research studies have increased exponentially over the years, highlighting the clinical significance and international interest in this disease. Hormonal imbalances have been linked to keratoconus, both clinically and experimentally, with both sexes affected. However, it is unclear how (molecular/cellular signaling) or when (age/disease stage(s)) those hormones affect the keratoconic cornea. Previous studies have categorized the human cornea as an extragonadal tissue, showing modulation of the gonadotropins, specifically luteinizing hormone (LH) and follicle-stimulating hormone (FSH). Studies herein provide new data (both in vitro and in vivo) to further delineate the role of hormones/gonadotropins in the keratoconus pathobiology, and propose the existence of a new axis named the Hypothalamic-Pituitary-Adrenal-Corneal (HPAC) axis.
Collapse
Affiliation(s)
- Dimitrios Karamichos
- North Texas Eye Research Institute, University of North Texas Health Science Center, 3430 Camp Bowie Blvd, Fort Worth, TX, 76107, USA; Department of Pharmaceutical Sciences, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA; Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA.
| | - Paulina Escandon
- North Texas Eye Research Institute, University of North Texas Health Science Center, 3430 Camp Bowie Blvd, Fort Worth, TX, 76107, USA; Department of Pharmaceutical Sciences, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA
| | - Brenda Vasini
- North Texas Eye Research Institute, University of North Texas Health Science Center, 3430 Camp Bowie Blvd, Fort Worth, TX, 76107, USA; Department of Pharmaceutical Sciences, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA
| | - Sarah E Nicholas
- North Texas Eye Research Institute, University of North Texas Health Science Center, 3430 Camp Bowie Blvd, Fort Worth, TX, 76107, USA; Department of Pharmaceutical Sciences, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA
| | - Lyly Van
- University of Oklahoma Health Sciences Center, 940 Stanton L Young, Oklahoma City, OK, USA; Department of Ophthalmology, Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Deanna H Dang
- College of Medicine, University of Oklahoma Health Sciences Center, 940 Stanton L Young, Oklahoma City, OK, USA
| | - Rebecca L Cunningham
- Department of Pharmaceutical Sciences, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA
| | - Kamran M Riaz
- Department of Ophthalmology, Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
16
|
Grisé KN, Coles BLK, Bautista NX, van der Kooy D. Activation of adult mammalian retinal stem cells in vivo via antagonism of BMP and sFRP2. Stem Cell Res Ther 2021; 12:560. [PMID: 34717744 PMCID: PMC8557620 DOI: 10.1186/s13287-021-02630-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 10/17/2021] [Indexed: 11/15/2022] Open
Abstract
Background The adult mammalian retina does not have the capacity to regenerate cells lost due to damage or disease. Therefore, retinal injuries and blinding diseases result in irreversible vision loss. However, retinal stem cells (RSCs), which participate in retinogenesis during development, persist in a quiescent state in the ciliary epithelium (CE) of the adult mammalian eye. Moreover, RSCs retain the ability to generate all retinal cell types when cultured in vitro, including photoreceptors. Therefore, it may be possible to activate endogenous RSCs to induce retinal neurogenesis in vivo and restore vision in the adult mammalian eye. Methods To investigate if endogenous RSCs can be activated, we performed combinatorial intravitreal injections of antagonists to BMP and sFRP2 proteins (two proposed mediators of RSC quiescence in vivo), with or without growth factors FGF and Insulin. We also investigated the effects of chemically-induced N-methyl-N-Nitrosourea (MNU) retinal degeneration on RSC activation, both alone and in combination withthe injected factors. Further, we employed inducible Msx1-CreERT2 genetic lineage labeling of the CE followed by stimulation paradigms to determine if activated endogenous RSCs could migrate into the retina and differentiate into retinal neurons. Results We found that in vivo antagonism of BMP and sFRP2 proteins induced CE cells in the RSC niche to proliferate and expanded the RSC population. BMP and sFRP2 antagonism also enhanced CE cell proliferation in response to exogenous growth factor stimulation and MNU-induced retinal degeneration. Furthermore, Msx1-CreERT2 genetic lineage tracing revealed that CE cells migrated into the retina following stimulation and/or injury, where they expressed markers of mature photoreceptors and retinal ganglion cells. Conclusions Together, these results indicate that endogenous adult mammalian RSCs may have latent regenerative potential that can be activated by modulating the RSC niche and hold promise as a means for endogenous retinal cell therapy to repair the retina and improve vision. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02630-0.
Collapse
Affiliation(s)
- Kenneth N Grisé
- Department of Molecular Genetics, University of Toronto, Donnelly Centre Rm 1110, 160 College Street, Toronto, ON, M5S 3E1, Canada.
| | - Brenda L K Coles
- Department of Molecular Genetics, University of Toronto, Donnelly Centre Rm 1110, 160 College Street, Toronto, ON, M5S 3E1, Canada
| | - Nelson X Bautista
- Department of Molecular Genetics, University of Toronto, Donnelly Centre Rm 1110, 160 College Street, Toronto, ON, M5S 3E1, Canada
| | - Derek van der Kooy
- Department of Molecular Genetics, University of Toronto, Donnelly Centre Rm 1110, 160 College Street, Toronto, ON, M5S 3E1, Canada.,Institute of Medical Science, University of Toronto, Toronto, ON, M5S 1A8, Canada
| |
Collapse
|
17
|
Owji AP, Kittredge A, Zhang Y, Yang T. Structure and Function of the Bestrophin family of calcium-activated chloride channels. Channels (Austin) 2021; 15:604-623. [PMID: 34612806 PMCID: PMC8496536 DOI: 10.1080/19336950.2021.1981625] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
Bestrophins are a family of calcium-activated chloride channels (CaCCs) with relevance to human physiology and a myriad of eye diseases termed "bestrophinopathies". Since the identification of bestrophins as CaCCs nearly two decades ago, extensive studies from electrophysiological and structural biology perspectives have sought to define their key channel features including calcium sensing, gating, inactivation, and anion selectivity. The initial X-ray crystallography studies on the prokaryotic homolog of Best1, Klebsiella pneumoniae (KpBest), and the Best1 homolog from Gallus gallus (chicken Best1, cBest1), laid the foundational groundwork for establishing the architecture of Best1. Recent progress utilizing single-particle cryogenic electron microscopy has further elucidated the molecular mechanism of gating in cBest1 and, separately, the structure of Best2 from Bos taurus (bovine Best2, bBest2). Meanwhile, whole-cell patch clamp, planar lipid bilayer, and other electrophysiologic analyses using these models as well as the human Best1 (hBest1) have provided ample evidence describing the functional properties of the bestrophin channels. This review seeks to consolidate these structural and functional results to paint a broad picture of the underlying mechanisms comprising the bestrophin family's structure-function relationship.
Collapse
Affiliation(s)
- Aaron P Owji
- Department of Pharmacology, Columbia University, NY, USA
| | - Alec Kittredge
- Department of Pharmacology, Columbia University, NY, USA
| | - Yu Zhang
- Department of Ophthalmology, Columbia University, NY, USA
| | - Tingting Yang
- Department of Ophthalmology, Columbia University, NY, USA
| |
Collapse
|
18
|
Hidalgo-Alvarez V, Dhowre HS, Kingston OA, Sheridan CM, Levis HJ. Biofabrication of Artificial Stem Cell Niches in the Anterior Ocular Segment. Bioengineering (Basel) 2021; 8:135. [PMID: 34677208 PMCID: PMC8533470 DOI: 10.3390/bioengineering8100135] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/27/2021] [Accepted: 09/27/2021] [Indexed: 11/16/2022] Open
Abstract
The anterior segment of the eye is a complex set of structures that collectively act to maintain the integrity of the globe and direct light towards the posteriorly located retina. The eye is exposed to numerous physical and environmental insults such as infection, UV radiation, physical or chemical injuries. Loss of transparency to the cornea or lens (cataract) and dysfunctional regulation of intra ocular pressure (glaucoma) are leading causes of worldwide blindness. Whilst traditional therapeutic approaches can improve vision, their effect often fails to control the multiple pathological events that lead to long-term vision loss. Regenerative medicine approaches in the eye have already had success with ocular stem cell therapy and ex vivo production of cornea and conjunctival tissue for transplant recovering patients' vision. However, advancements are required to increase the efficacy of these as well as develop other ocular cell therapies. One of the most important challenges that determines the success of regenerative approaches is the preservation of the stem cell properties during expansion culture in vitro. To achieve this, the environment must provide the physical, chemical and biological factors that ensure the maintenance of their undifferentiated state, as well as their proliferative capacity. This is likely to be accomplished by replicating the natural stem cell niche in vitro. Due to the complex nature of the cell microenvironment, the creation of such artificial niches requires the use of bioengineering techniques which can replicate the physico-chemical properties and the dynamic cell-extracellular matrix interactions that maintain the stem cell phenotype. This review discusses the progress made in the replication of stem cell niches from the anterior ocular segment by using bioengineering approaches and their therapeutic implications.
Collapse
Affiliation(s)
- Veronica Hidalgo-Alvarez
- Institute of Biological Chemistry, Biophysics and Bioengineering, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh EH14 4AS, UK
| | - Hala S. Dhowre
- Department of Eye and Vision Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, UK; (H.S.D.); (O.A.K.)
| | - Olivia A. Kingston
- Department of Eye and Vision Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, UK; (H.S.D.); (O.A.K.)
| | - Carl M. Sheridan
- Department of Eye and Vision Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, UK; (H.S.D.); (O.A.K.)
| | - Hannah J. Levis
- Department of Eye and Vision Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, UK; (H.S.D.); (O.A.K.)
| |
Collapse
|
19
|
Kattar A, Concheiro A, Alvarez-Lorenzo C. Diabetic eye: associated diseases, drugs in clinic, and role of self-assembled carriers in topical treatment. Expert Opin Drug Deliv 2021; 18:1589-1607. [PMID: 34253138 DOI: 10.1080/17425247.2021.1953466] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Introduction: Diabetes is a pandemic disease that causes relevant ocular pathologies. Diabetic retinopathy, macular edema, cataracts, glaucoma, or keratopathy strongly impact the quality of life of the patients. In addition to glycemic control, intense research is devoted to finding more efficient ocular drugs and improved delivery systems that can overcome eye barriers. Areas covered: The aim of this review is to revisit first the role of diabetes in the development of chronic eye diseases. Then, commercially available drugs and new candidates in clinical trials are tackled together with the pros and cons of their administration routes. Subsequent sections deal with self-assembled drug carriers suitable for eye instillation combining patient-friendly administration with high ocular bioavailability. Performance of topically administered polymeric micelles, liposomes, and niosomes for the management of diabetic eye diseases is analyzed in the light of ex vivo and in vivo results and outcomes of clinical trials. Expert opinion: Self-assembled carriers are being shown useful for efficient delivery of not only a variety of small drugs but also macromolecules (e.g. antibodies) and genes. Successful design of drug carriers may offer alternatives to intraocular injections and improve the treatment of both anterior and posterior segments diabetic eye diseases.
Collapse
Affiliation(s)
- Axel Kattar
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma Group (GI-1645), Facultad de Farmacia and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Angel Concheiro
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma Group (GI-1645), Facultad de Farmacia and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Carmen Alvarez-Lorenzo
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma Group (GI-1645), Facultad de Farmacia and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| |
Collapse
|
20
|
Choi Y, Jung K, Kim HJ, Chun J, Ahn M, Jee Y, Ko HJ, Moon C, Matsuda H, Tanaka A, Kim J, Shin T. Attenuation of Experimental Autoimmune Uveitis in Lewis Rats by Betaine. Exp Neurobiol 2021; 30:308-317. [PMID: 34483144 PMCID: PMC8424381 DOI: 10.5607/en21011] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 08/11/2021] [Accepted: 08/12/2021] [Indexed: 12/15/2022] Open
Abstract
Experimental autoimmune uveitis (EAU) is an animal model of human autoimmune uveitis that is characterized by the infiltration of autoimmune T cells with concurrent increases in pro-inflammatory cytokines and reactive oxygen species. This study aimed to assess whether betaine regulates the progression of EAU in Lewis rats. EAU was induced via immunization with the interphotoreceptor retinoid-binding protein (IRBP) and oral administration of either a vehicle or betaine (100 mg/kg) for 9 consecutive days. Spleens, blood, and retinas were sampled from the experimental rats at the time of sacrifice and used for the T cell proliferation assay, serological analysis, real-time polymerase chain reaction, and immunohistochemistry. The T cell proliferation assay revealed that betaine had little effect on the proliferation of splenic T cells against the IRBP antigen in an in vitro assay on day 9 post-immunization. The serological analysis showed that the level of serum superoxide dismutase increased in the betaine-treated group compared with that in the vehicle-treated group. The anti-inflammatory effect of betaine was confirmed by the downregulation of pro-inflammation-related molecules, including vascular cell adhesion molecule 1 and interleukin-1β in the retinas of rats with EAU. The histopathological findings agreed with those of ionized calcium-binding adaptor molecule 1 immunohistochemistry, further verifying that inflammation in the retina and ciliary bodies was significantly suppressed in the betaine-treated group compared with the vehicle-treated group. Results of the present study suggest that betaine is involved in mitigating EAU through anti-oxidation and anti-inflammatory activities.
Collapse
Affiliation(s)
- Yuna Choi
- College of Veterinary Medicine and Veterinary Medical Research Institute, Jeju National University, Jeju 63243, Korea
| | - Kyungsook Jung
- Functional Biomaterials Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup 56212, Korea
| | - Hyo Jin Kim
- Department of Food Bioengineering, Jeju National University, Jeju 63243, Korea
| | - Jiyoon Chun
- College of Veterinary Medicine and Veterinary Medical Research Institute, Jeju National University, Jeju 63243, Korea
| | - Meejung Ahn
- Department of Animal Science, College of Life Science, Sangji University, Wonju 26339, Korea
| | - Youngheun Jee
- College of Veterinary Medicine and Veterinary Medical Research Institute, Jeju National University, Jeju 63243, Korea
| | - Hyun Ju Ko
- College of Veterinary Medicine and Veterinary Medical Research Institute, Jeju National University, Jeju 63243, Korea
| | - Changjong Moon
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Gwangju 61186, Korea
| | - Hiroshi Matsuda
- Laboratory of Veterinary Molecular Pathology and Therapeutics, Division of Animal Life Science, Graduate School, Institute of Agriculture, Tokyo University of Agriculture and Technology, Tokyo 183-850
| | - Akane Tanaka
- Laboratory of Veterinary Molecular Pathology and Therapeutics, Division of Animal Life Science, Graduate School, Institute of Agriculture, Tokyo University of Agriculture and Technology, Tokyo 183-850
| | - Jeongtae Kim
- Department of Anatomy, Kosin University College of Medicine, Busan 49267, Korea
| | - Taekyun Shin
- College of Veterinary Medicine and Veterinary Medical Research Institute, Jeju National University, Jeju 63243, Korea
| |
Collapse
|
21
|
Schneider-Futschik EK, Reyes-Ortega F. Advantages and Disadvantages of Using Magnetic Nanoparticles for the Treatment of Complicated Ocular Disorders. Pharmaceutics 2021; 13:1157. [PMID: 34452117 PMCID: PMC8400382 DOI: 10.3390/pharmaceutics13081157] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/20/2021] [Accepted: 07/22/2021] [Indexed: 01/01/2023] Open
Abstract
Nanomaterials provide enormous opportunities to overcome the limitations of conventional ocular delivery systems, such as low therapeutic efficacy, side effects due to the systemic exposure, or invasive surgery. Apart from the more common ocular disorders, there are some genetic diseases, such as cystic fibrosis, that develop ocular disorders as secondary effects as long as the disease progresses. These patients are more difficult to be pharmacologically treated using conventional drug routes (topically, systemic), since specific pharmacological formulations can be incompatible, display increased toxicity, or their therapeutic efficacy decreases with the administration of different kind of chemical molecules. Magnetic nanoparticles can be used as potent drug carriers and magnetic hyperthermia agents due to their response to an external magnetic field. Drugs can be concentrated in the target point, limiting the damage to other tissues. The other advantage of these magnetic nanoparticles is that they can act as magnetic resonance imaging agents, allowing the detection of the exact location of the disease. However, there are some drawbacks related to their use in drug delivery, such as the limitation to maintain efficacy in the target organ once the magnetic field is removed from outside. Another disadvantage is the difficulty in maintaining the therapeutic action in three dimensions inside the human body. This review summarizes all the application possibilities related to magnetic nanoparticles in ocular diseases.
Collapse
Affiliation(s)
- Elena K. Schneider-Futschik
- Department of Biochemistry & Pharmacology, Faculty of Medicine, School of Biomedical Sciences, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC 3010, Australia;
| | - Felisa Reyes-Ortega
- Visual Quality Research Group, Department of Ophthalmology, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofía University Hospital and University of Cordoba, 14004 Cordoba, Spain
| |
Collapse
|
22
|
Escandon P, Vasini B, Whelchel AE, Nicholas SE, Matlock HG, Ma JX, Karamichos D. The role of peroxisome proliferator-activated receptors in healthy and diseased eyes. Exp Eye Res 2021; 208:108617. [PMID: 34010603 PMCID: PMC8594540 DOI: 10.1016/j.exer.2021.108617] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 05/03/2021] [Accepted: 05/05/2021] [Indexed: 12/23/2022]
Abstract
Peroxisome Proliferator-Activated Receptors (PPARs) are a family of nuclear receptors that play essential roles in modulating cell differentiation, inflammation, and metabolism. Three subtypes of PPARs are known: PPAR-alpha (PPARα), PPAR-gamma (PPARγ), and PPAR-beta/delta (PPARβ/δ). PPARα activation reduces lipid levels and regulates energy homeostasis, activation of PPARγ results in regulation of adipogenesis, and PPARβ/δ activation increases fatty acid metabolism and lipolysis. PPARs are linked to various diseases, including but not limited to diabetes, non-alcoholic fatty liver disease, glaucoma and atherosclerosis. In the past decade, numerous studies have assessed the functional properties of PPARs in the eye and key PPAR mechanisms have been discovered, particularly regarding the retina and cornea. PPARγ and PPARα are well established in their functions in ocular homeostasis regarding neuroprotection, neovascularization, and inflammation, whereas PPARβ/δ isoform function remains understudied. Naturally, studies on PPAR agonists and antagonists, associated with ocular pathology, have also gained traction with the development of PPAR synthetic ligands. Studies on PPARs has significantly influenced novel therapeutics for diabetic eye disease, ocular neuropathy, dry eye, and age-related macular degeneration (AMD). In this review, therapeutic potentials and implications will be highlighted, as well as reported adverse effects. Further investigations are necessary before any of the PPARs ligands can be utilized, in the clinics, to treat eye diseases. Future research on the prominent role of PPARs will help unravel the complex mechanisms involved in order to prevent and treat ocular diseases.
Collapse
Affiliation(s)
- Paulina Escandon
- North Texas Eye Research Institute, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA; Department of Pharmaceutical Sciences, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA
| | - Brenda Vasini
- North Texas Eye Research Institute, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA; Department of Pharmaceutical Sciences, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA
| | - Amy E Whelchel
- Department of Physiology, University of Oklahoma Health Sciences Center, 940 Stanton L Young, Oklahoma City, OK, USA
| | - Sarah E Nicholas
- North Texas Eye Research Institute, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA; Department of Pharmaceutical Sciences, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA
| | - H Greg Matlock
- Department of Physiology, University of Oklahoma Health Sciences Center, 940 Stanton L Young, Oklahoma City, OK, USA
| | - Jian-Xing Ma
- Department of Physiology, University of Oklahoma Health Sciences Center, 940 Stanton L Young, Oklahoma City, OK, USA; Harold Hamm Oklahoma Diabetes Center, 1000 N Lincoln Blvd, Oklahoma City, OK, USA
| | - Dimitrios Karamichos
- North Texas Eye Research Institute, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA; Department of Pharmaceutical Sciences, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA; Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA.
| |
Collapse
|
23
|
Frasson LT, Dalmaso B, Akamine PS, Kimura ET, Hamassaki DE, Del Debbio CB. Let-7, Lin28 and Hmga2 Expression in Ciliary Epithelium and Retinal Progenitor Cells. Invest Ophthalmol Vis Sci 2021; 62:31. [PMID: 33749722 PMCID: PMC7991968 DOI: 10.1167/iovs.62.3.31] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 02/24/2021] [Indexed: 12/03/2022] Open
Abstract
Purpose Ciliary epithelium (CE) of adult mammalian eyes contains quiescent retinal progenitor/stem cells that generate neurospheres in vitro and differentiate into retinal neurons. This ability doesn't evolve efficiently probably because of regulatory mechanisms, such as microRNAs (miRNAs) that control pluripotent, progenitor, and differentiation genes. Here we investigate the presence of Let-7 miRNAs and its regulator and target, Lin28 and Hmga2, in CE cells from neurospheres, newborns, and adult tissues. Methods Newborn and adult rats CE cells were dissected into pigmented and nonpigmented epithelium (PE and NPE). Newborn PE cells were cultured with growth factors to form neurospheres and we analyzed Let-7, Lin28a, and Hmga2 expression. During the neurospheres formation, we added chemically modified single-stranded oligonucleotides designed to bind and inhibit or mimic endogenous mature Let-7b and Let-7c. After seven days in culture, we analyzed neurospheres size, number and expression of Let-7, Lin28, and Hmga2. Results Let-7 miRNAs were expressed at low rates in newborn CE cells with significant increase in adult tissues, with higher levels on NPE cells, that does not present the stem cells reprogramming ability. The Lin28a and Hmga2 protein and transcripts were more expressed in newborns than adults cells, opposed to Let-7. Neurospheres presented higher Lin28 and Hmga2 expression than newborn and adult, but similar Let-7 than newborns. Let-7b inhibitor upregulated Hmga2 expression, whereas Let-7c mimics upregulated Lin28 and downregulated Hmga2. Conclusions This study shows the dynamic of Lin28-Let-7-Hmga regulatory axis in CE cells. These components may develop different roles during neurospheres formation and postnatal CE cells.
Collapse
Affiliation(s)
- Lorena Teixeira Frasson
- Department of Cell Biology and Development, Biomedical Sciences Institute, University of Sao Paulo, Sao Paulo, Brazil
| | - Barbara Dalmaso
- Department of Cell Biology and Development, Biomedical Sciences Institute, University of Sao Paulo, Sao Paulo, Brazil
| | - Priscilla Sayami Akamine
- Department of Cell Biology and Development, Biomedical Sciences Institute, University of Sao Paulo, Sao Paulo, Brazil
| | - Edna Teruko Kimura
- Department of Cell Biology and Development, Biomedical Sciences Institute, University of Sao Paulo, Sao Paulo, Brazil
| | - Dânia Emi Hamassaki
- Department of Cell Biology and Development, Biomedical Sciences Institute, University of Sao Paulo, Sao Paulo, Brazil
| | - Carolina Beltrame Del Debbio
- Department of Cell Biology and Development, Biomedical Sciences Institute, University of Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
24
|
Pang J, Le L, Zhou Y, Tu R, Hou Q, Tsuchiya D, Thomas N, Wang Y, Yu Z, Alexander R, Thexton M, Lewis B, Corbin T, Durnin M, Li H, Ashery-Padan R, Yan D, Xie T. NOTCH Signaling Controls Ciliary Body Morphogenesis and Secretion by Directly Regulating Nectin Protein Expression. Cell Rep 2021; 34:108603. [PMID: 33440163 DOI: 10.1016/j.celrep.2020.108603] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 11/20/2020] [Accepted: 12/14/2020] [Indexed: 11/29/2022] Open
Abstract
Anterior segment dysgenesis is often associated with cornea diseases, cataracts, and glaucoma. In the anterior segment, the ciliary body (CB) containing inner and outer ciliary epithelia (ICE and OCE) secretes aqueous humor that maintains intraocular pressure (IOP). However, CB development and function remain poorly understood. Here, this study shows that NOTCH signaling in the CB maintains the vitreous, IOP, and eye structures by regulating CB morphogenesis, aqueous humor secretion, and vitreous protein expression. Notch2 and Notch3 function via RBPJ in the CB to control ICE-OCE adhesion, CB morphogenesis, aqueous humor secretion, and protein expression, thus maintaining IOP and eye structures. Mechanistically, NOTCH signaling transcriptionally controls Nectin1 expression in the OCE to promote cell adhesion for driving CB morphogenesis and to directly stabilize Cx43 for controlling aqueous humor secretion. Finally, NOTCH signaling directly controls vitreous protein secretion in the ICE. Therefore, this study provides important insight into CB functions and involvement in eye diseases.
Collapse
Affiliation(s)
- Ji Pang
- Stowers Institute for Medical Research, 1000 East 50(th) Street, Kansas City, MO 64110, USA; School of Chemistry and Chemical Engineering, State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Liang Le
- Stowers Institute for Medical Research, 1000 East 50(th) Street, Kansas City, MO 64110, USA
| | - Yi Zhou
- Stowers Institute for Medical Research, 1000 East 50(th) Street, Kansas City, MO 64110, USA; Department of Anatomy and Cell Biology, University of Kansas School of Medicine, 3901 Rainbow Blvd, Kansas City, KS 66160, USA
| | - Renjun Tu
- Stowers Institute for Medical Research, 1000 East 50(th) Street, Kansas City, MO 64110, USA
| | - Qiang Hou
- Stowers Institute for Medical Research, 1000 East 50(th) Street, Kansas City, MO 64110, USA; State Key Laboratory and Key Laboratory of Vision Science, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Dai Tsuchiya
- Stowers Institute for Medical Research, 1000 East 50(th) Street, Kansas City, MO 64110, USA
| | - Nancy Thomas
- Stowers Institute for Medical Research, 1000 East 50(th) Street, Kansas City, MO 64110, USA
| | - Yongfu Wang
- Stowers Institute for Medical Research, 1000 East 50(th) Street, Kansas City, MO 64110, USA
| | - Zulin Yu
- Stowers Institute for Medical Research, 1000 East 50(th) Street, Kansas City, MO 64110, USA
| | - Richard Alexander
- Stowers Institute for Medical Research, 1000 East 50(th) Street, Kansas City, MO 64110, USA
| | - Marina Thexton
- Stowers Institute for Medical Research, 1000 East 50(th) Street, Kansas City, MO 64110, USA
| | - Brandy Lewis
- Stowers Institute for Medical Research, 1000 East 50(th) Street, Kansas City, MO 64110, USA
| | - Timothy Corbin
- Stowers Institute for Medical Research, 1000 East 50(th) Street, Kansas City, MO 64110, USA
| | - Michael Durnin
- Stowers Institute for Medical Research, 1000 East 50(th) Street, Kansas City, MO 64110, USA
| | - Hua Li
- Stowers Institute for Medical Research, 1000 East 50(th) Street, Kansas City, MO 64110, USA
| | - Ruth Ashery-Padan
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Deyue Yan
- School of Chemistry and Chemical Engineering, State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Ting Xie
- Stowers Institute for Medical Research, 1000 East 50(th) Street, Kansas City, MO 64110, USA; Department of Anatomy and Cell Biology, University of Kansas School of Medicine, 3901 Rainbow Blvd, Kansas City, KS 66160, USA.
| |
Collapse
|
25
|
Lincke JB, Keller S, Amaral J, Zinkernagel MS, Schuerch K. Ciliary body length revisited by anterior segment optical coherence tomography: implications for safe access to the pars plana for intravitreal injections. Graefes Arch Clin Exp Ophthalmol 2020; 259:1435-1441. [PMID: 33074373 PMCID: PMC8166736 DOI: 10.1007/s00417-020-04967-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 09/30/2020] [Accepted: 10/02/2020] [Indexed: 11/27/2022] Open
Abstract
Purpose To investigate the dependence of the ciliary body length (CBL) on the axial length (AL) and to draw conclusions on implications regarding safe pars plana access for intravitreal injections and vitreoretinal surgery. Methods A total of 200 individuals (mean age 42 years, SD ± 15.4) were enrolled in the study. Objective refraction and AL were obtained. Spherical equivalent (SE) was calculated. Anterior segment optical coherence tomography (ASOCT) was used to image and measure the CBL. Results The mean SE was − 1.64 diopters (SD ± 3.15, range − 14.5 to + 9 diopters) and the mean AL was 24.19 mm (SD ± 1.65, range 19.8–32.2 mm). There was a significant correlation between SE and AL (r2 = 0.62, p < 0.0001). Mean CBL correlated significantly with age (r2 = 0.11, p < 0.0001), AL (r2 = 0.23, p < 0.0001) and SE (r2 = 0.25, p < 0.0001). The mean CBL was 3351 μm (SD ± 459, range 2184–4451 μm). Three separate groups were defined by their AL with a normal AL group (AL 22.5 to 25 mm), a short AL group (AL < 22.5 mm) and a long AL group (AL > 25 mm). The mean CBL in the normal AL group was 3311 μm (SD ± 427), in the short AL group 2936 μm (SD ± 335) and in the long AL group 3715 μm (SD ± 365), and differed significantly (p < 0.0001) when compared. Conclusion For interventions requiring pars plana access (as an intravitreal injection or vitreoretinal surgery), an incision distance of 3.5–4.0 mm posterior to the limbus is recommended. In our research, however, a difference of 0.77 mm in mean CBL between the group with short AL and the group with long AL is demonstrated, implying that the mean CBL in very short and very long eyes differs significantly. These findings suggest that the AL should be taken into account for pars plana access and that it would be advisable to prefer the shorter or longer recommended distance (3.5 and 4.0 mm, respectively) from the limbus, which correlates with the AL. If AL is > 25 mm, a distance of 4.0 mm from the limbus should be chosen; and if AL is < 22.5 mm, a distance of 3.5 mm seems adequate. Trial registration number and date NCT00564291, 27 Nov 2007![]()
Collapse
Affiliation(s)
- Joel-Benjamin Lincke
- Department of Ophthalmology, Inselspital, University Hospital of Bern, Bern, Switzerland.
| | - Salome Keller
- Department of Ophthalmology, Inselspital, University Hospital of Bern, Bern, Switzerland
| | - Joao Amaral
- Department of Ophthalmology, Inselspital, University Hospital of Bern, Bern, Switzerland
| | - Martin S Zinkernagel
- Department of Ophthalmology, Inselspital, University Hospital of Bern, Bern, Switzerland
| | - Kaspar Schuerch
- Department of Ophthalmology, Inselspital, University Hospital of Bern, Bern, Switzerland
| |
Collapse
|
26
|
Lu R, Soden PA, Lee E. Tissue-Engineered Models for Glaucoma Research. MICROMACHINES 2020; 11:mi11060612. [PMID: 32599818 PMCID: PMC7345325 DOI: 10.3390/mi11060612] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 06/21/2020] [Accepted: 06/22/2020] [Indexed: 12/20/2022]
Abstract
Glaucoma is a group of optic neuropathies characterized by the progressive degeneration of retinal ganglion cells (RGCs). Patients with glaucoma generally experience elevations in intraocular pressure (IOP), followed by RGC death, peripheral vision loss and eventually blindness. However, despite the substantial economic and health-related impact of glaucoma-related morbidity worldwide, the surgical and pharmacological management of glaucoma is still limited to maintaining IOP within a normal range. This is in large part because the underlying molecular and biophysical mechanisms by which glaucomatous changes occur are still unclear. In the present review article, we describe current tissue-engineered models of the intraocular space that aim to advance the state of glaucoma research. Specifically, we critically evaluate and compare both 2D and 3D-culture models of the trabecular meshwork and nerve fiber layer, both of which are key players in glaucoma pathophysiology. Finally, we point out the need for novel organ-on-a-chip models of glaucoma that functionally integrate currently available 3D models of the retina and the trabecular outflow pathway.
Collapse
Affiliation(s)
- Renhao Lu
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA;
| | - Paul A. Soden
- College of Human Ecology, Cornell University, Ithaca, NY 14853, USA;
| | - Esak Lee
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA;
- Correspondence: ; Tel.: +1-607-255-8491
| |
Collapse
|
27
|
Dobbs NW, Budak MJ, White RD, Zealley IA. MR-Eye: High-Resolution Microscopy Coil MRI for the Assessment of the Orbit and Periorbital Structures, Part 1: Technique and Anatomy. AJNR Am J Neuroradiol 2020; 41:947-950. [PMID: 32241775 DOI: 10.3174/ajnr.a6495] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 01/21/2020] [Indexed: 11/07/2022]
Abstract
Microscopy coil MR imaging of the orbits has been described previously as a technique for anatomic depiction. In the first part of this 2-part series, the improvement in spatial resolution that the technique offers compared with conventional MR imaging of the orbits is demonstrated. We provide a guide to implementing the technique, sharing pearls and pitfalls gleaned from our own practice to make implementation of microscopy coil MR imaging at your own center easy. As a quick reference guide to the small-scale structures encountered when reading the studies, a short anatomy section is included, which doubles as a showcase for the high-quality imaging that can be obtained. In the second part, our experience of microscopy coil MR imaging in day-to-day clinical practice takes it far beyond being a useful anatomic educational tool. Through a series of interesting cases, we highlight the added benefit of microscopy coil MR imaging compared with standard orbital MR imaging.
Collapse
Affiliation(s)
- N W Dobbs
- From the Department of Clinical Neuroscience (N.W.D.), Western General Hospital, Edinburgh, United Kingdom
| | - M J Budak
- Qscan Radiology Clinics (M.J.B.), Gold Coast, Australia
| | - R D White
- Department of Clinical Radiology (R.D.W.), University Hospital of Wales, Cardiff, United Kingdom
| | - I A Zealley
- Department of Clinical Radiology (I.A.Z.), Ninewells Hospital, Dundee, United Kingdom
| |
Collapse
|
28
|
Alkozi HA, Navarro G, Aguinaga D, Reyes-Resina I, Sanchez-Naves J, Pérez de Lara MJ, Franco R, Pintor J. Adreno-melatonin receptor complexes control ion homeostasis and intraocular pressure - their disruption contributes to hypertensive glaucoma. Br J Pharmacol 2020; 177:2090-2105. [PMID: 31901203 DOI: 10.1111/bph.14971] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 12/05/2019] [Accepted: 12/09/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND AND PURPOSE Often, glaucoma presents with elevated eye hydrostatic pressure, which is regulated by endogenous melatonin. Phenylephrine increases cytoplasmic [Ca2+ ], via α1 -adrenoceptor activation, that is detrimental in glaucoma. The aims of this study were (a) to elucidate the role of melatonin receptors in humour production and intraocular pressure (IOP) maintenance and (b) to identify glaucoma-relevant melatonin-adrenoceptor interactions. EXPERIMENTAL APPROACH Biophysical and proximity ligation assays were performed to identify interactions in heterologous expression systems, in cell lines and in human eyes. Gs /Gi /Gq signalling was investigated in these systems and in cells producing aqueous humour. IOP was determined in a mice model of glaucoma. Retinography and topically pharmacological treatment were performed in control and in glaucomatous mice. KEY RESULTS α1 -adreno- and melatonin receptors form functional complexes in which the C-terminal tail of the adrenoceptor plays a role. Remarkably, activation of α1 -adrenoceptors in these complexes did not lead to cytosolic Ca2+ increases, suggesting Gs instead of Gq coupling is involved. The number of these complexes significantly decreased in models of glaucoma and, importantly, in human samples from glaucoma patients. This has led to hypothesize that melatonin, a hypotensive agent, plus blockade of α1 -adrenoceptors could normalize pressure in glaucoma. Remarkably, co-instillation of melatonin and prazosin, an α1 -adrenoceptor antagonist, resulted in long-term decreases in IOP in a well-established animal model of glaucoma. CONCLUSIONS AND IMPLICATIONS The findings are instrumental to understand the physiological function of melatonin in the eye and its potential to address eye pathologies by targeting melatonin receptors and their complexes.
Collapse
Affiliation(s)
- Hanan Awad Alkozi
- Department of Biochemistry and Molecular Biology, Faculty of Optics and Optometry, University Complutense of Madrid, Madrid, Spain
| | - Gemma Navarro
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CiberNed), Instituto de Salud Carlos III, Madrid, Spain.,Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Universitat de Barcelona, Barcelona, Spain
| | - David Aguinaga
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CiberNed), Instituto de Salud Carlos III, Madrid, Spain.,Department of Biochemistry and Molecular Biomedicine, School of Biology, Universitat de Barcelona, Barcelona, Spain
| | - Irene Reyes-Resina
- Department of Biochemistry and Molecular Biomedicine, School of Biology, Universitat de Barcelona, Barcelona, Spain.,Research Group Neuroplasticity, Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Juan Sanchez-Naves
- Department of Ophthalmology, Balearic Islands Institute of Ophthalmology, Palma de Mallorca, Spain
| | - Maria J Pérez de Lara
- Department of Biochemistry and Molecular Biology, Faculty of Optics and Optometry, University Complutense of Madrid, Madrid, Spain
| | - Rafael Franco
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CiberNed), Instituto de Salud Carlos III, Madrid, Spain.,Department of Biochemistry and Molecular Biomedicine, School of Biology, Universitat de Barcelona, Barcelona, Spain
| | - Jesus Pintor
- Department of Biochemistry and Molecular Biology, Faculty of Optics and Optometry, University Complutense of Madrid, Madrid, Spain
| |
Collapse
|
29
|
Ocular Hypotonia and Transient Decrease of Vision as a Consequence of Exposure to a Common Toad Poison. Case Rep Ophthalmol Med 2020; 2020:2983947. [PMID: 32015920 PMCID: PMC6988675 DOI: 10.1155/2020/2983947] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 01/07/2020] [Indexed: 11/24/2022] Open
Abstract
The common toad produces venom (bufotoxin) that is produced in the parotid gland of the toad as well as in the skin. This toxic compound is a potent inhibitor of Na+/K+-ATPase activity. Physiological effects of bufotoxin are similar to those of digitalis and cause increased heart rate and muscle contractions. Ocular toxicity was described. A 67-year-old female patient was admitted to the emergency service because of sudden vision loss and a burning sensation in both eyes after she had been exposed to the poison of a toad. Slit lamp examination showed conjunctival hyperaemia and signs of ocular hypotonia. Topical antibiotic treatment was administered, and after 24 hours, corneal oedema and ocular hypotonia were in remission. Inhibition of Na+/K+-ATPase is a well-known effect of the toad venom. Na+/K+-ATPase is a part of corneal endothelial cells, ciliary body, and iris, and its inhibition caused by exposure to bufadienolides induces corneal dysfunction, decreased vision, and ocular hypotonia. Effects of bufadienolides on the decrease of ocular pressure appear to be very strong, with quick action. This rarely described effect of the bufotoxin can be used as a basis for further research of toad venom and its pharmacological potential. Purpose. To present a case of a 67-year-old female patient who experienced a sudden decrease in vision after exposure to the poison from a common toad (Bufo bufo).
Collapse
|
30
|
Reyes-Resina I, Awad Alkozi H, del Ser-Badia A, Sánchez-Naves J, Lillo J, Jiménez J, Pintor J, Navarro G, Franco R. Expression of Melatonin and Dopamine D 3 Receptor Heteromers in Eye Ciliary Body Epithelial Cells and Negative Correlation with Ocular Hypertension. Cells 2020; 9:cells9010152. [PMID: 31936298 PMCID: PMC7016594 DOI: 10.3390/cells9010152] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 12/23/2019] [Accepted: 01/02/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Experiments in the late nineties showed an inverse relationship in the eye levels of melatonin and dopamine, thereby constituting an example of eye parameters that are prone to circadian variations. The underlying mechanisms are not known but these relevant molecules act via specific cell surface dopamine and melatonin receptors. This study investigated whether these receptors formed heteromers whose function impact on eye physiology. We performed biophysical assays to identify interactions in heterologous systems. Particular heteromer functionality was detected using Gi coupling, MAPK activation, and label-free assays. The expression of the heteroreceptor complexes was assessed using proximity ligation assays in cells producing the aqueous humor and human eye samples. Dopamine D3 receptors (D3Rs) were identified in eye ciliary body epithelial cells. We discovered heteromers formed by D3R and either MT1 (MT1R) or MT2 (MT2R) melatonin receptors. Heteromerization led to the blockade of D3R-Gi coupling and regulation of signaling to the MAPK pathway. Heteromer expression was negatively correlated with intraocular hypertension. CONCLUSIONS Heteromers likely mediate melatonin and dopamine actions in structures regulating intraocular pressure. Significant expression of D3R-MT1R and D3R-MT1R was associated with normotensive conditions, whereas expression diminished in a cell model of hypertension. A clear trend of expression reduction was observed in samples from glaucoma cases. The trend was marked but no statistical analysis was possible as the number of available eyes was 2.
Collapse
Affiliation(s)
- Irene Reyes-Resina
- Department of Biochemistry and Molecular Biomedicine, School of Biology, Universitat de Barcelona, 08028 Barcelona, Spain;
- Neuroplasticity Research Group, Leibniz Institute for Neurobiology, 39118 Magdeburg, Germany
- Centro de Investigación en Red, Enfermedades Neurodegenerativas, CiberNed, Instituto de Salud Carlos III, 28029 Madrid, Spain; (A.d.S.-B.); (J.J.)
- Correspondence: (I.R.-R.); (G.N.); or (R.F.); Tel.: +34-934021208 (I.R.-R. & G.N.)
| | - Hanan Awad Alkozi
- Department of Biochemistry and Molecular Biology, Faculty of Optics and Optometry, University Complutense of Madrid, 28037 Madrid, Spain; (H.A.A.); (J.P.)
| | - Anna del Ser-Badia
- Centro de Investigación en Red, Enfermedades Neurodegenerativas, CiberNed, Instituto de Salud Carlos III, 28029 Madrid, Spain; (A.d.S.-B.); (J.J.)
- Department de Bioquímica i Biologia Molecular, Institut de Neurociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, 08193 Barcelona, Spain
| | - Juan Sánchez-Naves
- Department of Ophthalmology, Balearic Islands Institute of Ophthalmology, 07013 Palma de Mallorca, Mallorca, Spain;
| | - Jaume Lillo
- Department of Biochemistry and Molecular Biomedicine, School of Biology, Universitat de Barcelona, 08028 Barcelona, Spain;
- Centro de Investigación en Red, Enfermedades Neurodegenerativas, CiberNed, Instituto de Salud Carlos III, 28029 Madrid, Spain; (A.d.S.-B.); (J.J.)
| | - Jasmina Jiménez
- Centro de Investigación en Red, Enfermedades Neurodegenerativas, CiberNed, Instituto de Salud Carlos III, 28029 Madrid, Spain; (A.d.S.-B.); (J.J.)
| | - Jesús Pintor
- Department of Biochemistry and Molecular Biology, Faculty of Optics and Optometry, University Complutense of Madrid, 28037 Madrid, Spain; (H.A.A.); (J.P.)
| | - Gemma Navarro
- Centro de Investigación en Red, Enfermedades Neurodegenerativas, CiberNed, Instituto de Salud Carlos III, 28029 Madrid, Spain; (A.d.S.-B.); (J.J.)
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Universitat de Barcelona, 08027 Barcelona, Spain
- Correspondence: (I.R.-R.); (G.N.); or (R.F.); Tel.: +34-934021208 (I.R.-R. & G.N.)
| | - Rafael Franco
- Centro de Investigación en Red, Enfermedades Neurodegenerativas, CiberNed, Instituto de Salud Carlos III, 28029 Madrid, Spain; (A.d.S.-B.); (J.J.)
- School of Chemistry, Universitat de Barcelona, 08028 Barcelona, Spain
- Correspondence: (I.R.-R.); (G.N.); or (R.F.); Tel.: +34-934021208 (I.R.-R. & G.N.)
| |
Collapse
|
31
|
Martis RM, Donaldson PJ, Li B, Middleditch M, Kallingappa PK, Lim JC. Mapping of the cystine-glutamate exchanger in the mouse eye: a role for xCT in controlling extracellular redox balance. Histochem Cell Biol 2019; 152:293-310. [PMID: 31396687 DOI: 10.1007/s00418-019-01805-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/24/2019] [Indexed: 12/13/2022]
Abstract
The cystine-glutamate exchanger (system xc-) is responsible for the exchange of extracellular cystine for intracellular glutamate. In this study, we mapped the expression of xCT, the light chain subunit of system xc- in the different tissues of 3-6-week-old mouse (C57BL/6J) eye and have used an xCT knockout mouse to verify labelling specificity. Moreover, using the xCT knockout mouse, we investigated whether xCT was involved in maintaining extracellular redox balance in the eye. xCT transcript and protein were present in the cornea, lens and retina of wild-type mice, but not knockout mice. xCT was localised to the corneal epithelium, and the lens epithelium and cortical fibre cells but was absent in the iris. xCT localisation could not be determined in the ciliary body or retina, since xCT labelling was also detected in the knockout indicating a lack of specificity of the xCT antibody in tissues of a neural origin. Intracellular cysteine and cystine concentrations were similar in the wild-type and xCT knockout mouse for the cornea, lens, and retina. While extracellular cysteine levels were similar between the plasma, aqueous humour, and vitreous humour of the wild-type and xCT knockout mouse, extracellular cystine levels in the plasma and aqueous were significantly elevated in the xCT knockout mouse relative to the wild type. This suggests that loss of xCT results in an increased oxidative environment, particularly within the anterior chamber of the eye in which the aqueous humour resides. How this oxidative shift impacts ocular tissues that interface with the aqueous humour over time will be the focus of future work.
Collapse
Affiliation(s)
- Renita M Martis
- Department of Physiology, School of Medical and Health Sciences, University of Auckland, Auckland, 1023, New Zealand.,School of Medical Sciences, University of Auckland, Auckland, New Zealand.,NZ National Eye Centre, University of Auckland, Auckland, New Zealand
| | - Paul J Donaldson
- Department of Physiology, School of Medical and Health Sciences, University of Auckland, Auckland, 1023, New Zealand.,School of Medical Sciences, University of Auckland, Auckland, New Zealand.,NZ National Eye Centre, University of Auckland, Auckland, New Zealand
| | - Bo Li
- Department of Physiology, School of Medical and Health Sciences, University of Auckland, Auckland, 1023, New Zealand.,School of Medical Sciences, University of Auckland, Auckland, New Zealand.,NZ National Eye Centre, University of Auckland, Auckland, New Zealand
| | - Martin Middleditch
- School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Prasanna K Kallingappa
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand.,School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Julie C Lim
- Department of Physiology, School of Medical and Health Sciences, University of Auckland, Auckland, 1023, New Zealand. .,School of Medical Sciences, University of Auckland, Auckland, New Zealand. .,NZ National Eye Centre, University of Auckland, Auckland, New Zealand.
| |
Collapse
|
32
|
Burgos J, Villanueva S, Ojeda M, Cornejo I, Cid LP, Sepúlveda FV. Kir7.1 inwardly rectifying K + channel is expressed in ciliary body non pigment epithelial cells and might contribute to intraocular pressure regulation. Exp Eye Res 2019; 186:107723. [PMID: 31319081 DOI: 10.1016/j.exer.2019.107723] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 06/26/2019] [Accepted: 07/10/2019] [Indexed: 02/08/2023]
Abstract
Inwardly rectifying K+ channel Kir7.1 is expressed in epithelia where it shares membrane localisation with the Na+/K+-pump. The ciliary body epithelium (CBE) of the eye is a determinant of intraocular pressure (IOP) through NaCl-driven fluid secretion of aqueous humour. In the present study we explored the presence Kir7.1 in this epithelium in the mouse and its possible functional role in the generation of IOP. Use heterozygous animals for total Kir7.1 knockout expressing β-galactosidase under the control of Kir7.1 promoter, identified the expression of Kir7.1 in non-pigmented epithelial cells of CBE. Using conditional, floxed knockout Kir7.1 mice as negative controls, we found Kir7.1 at the basolateral membrane of the same CBE cell layer. This was confirmed using a knockin mouse expressing the Kir7.1 protein tagged with a haemagglutinin epitope. Measurements using the conditional knockout mouse show only a minor effect of Kir7.1 inactivation on steady-state IOP. Transient increases in IOP in response to general anaesthetics, or to water injection, are absent or markedly curtailed in Kir7.1-deficient mice. These results suggest a role for Kir7.1 in IOP regulation through a possible modulation of aqueous humour production by the CBE non-pigmented epithelial cells. The location of Kir7.1 in the CBE, together with the effect of its removal on dynamic changes in IOP, point to a possible role of the channel as a leak pathway preventing cellular overload of K+ during the secretion process. Kir7.1 could be used as a potential therapeutic target in pathological conditions leading to elevated intraocular pressure.
Collapse
Affiliation(s)
- Johanna Burgos
- Centro de Estudios Científicos (CECs), Avenida Arturo Prat 514, Valdivia, Chile
| | - Sandra Villanueva
- Centro de Estudios Científicos (CECs), Avenida Arturo Prat 514, Valdivia, Chile; Universidad Austral de Chile, Valdivia, Chile
| | - Margarita Ojeda
- Centro de Estudios Científicos (CECs), Avenida Arturo Prat 514, Valdivia, Chile
| | - Isabel Cornejo
- Centro de Estudios Científicos (CECs), Avenida Arturo Prat 514, Valdivia, Chile
| | - L Pablo Cid
- Centro de Estudios Científicos (CECs), Avenida Arturo Prat 514, Valdivia, Chile
| | | |
Collapse
|
33
|
McWilliams TG, Prescott AR, Villarejo-Zori B, Ball G, Boya P, Ganley IG. A comparative map of macroautophagy and mitophagy in the vertebrate eye. Autophagy 2019; 15:1296-1308. [PMID: 30786807 PMCID: PMC6613837 DOI: 10.1080/15548627.2019.1580509] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 01/11/2019] [Accepted: 01/29/2019] [Indexed: 02/08/2023] Open
Abstract
Photoreception is pivotal to our experience and perception of the natural world; hence the eye is of prime importance for most vertebrate animals to sense light. Central to visual health is mitochondrial homeostasis, and the selective autophagic turnover of mitochondria (mitophagy) is predicted to play a key role here. Despite studies that link aberrant mitophagy to ocular dysfunction, little is known about the prevalence of basal mitophagy, or its relationship to general autophagy, in the visual system. In this study, we utilize the mito-QC mouse and a closely related general macroautophagy reporter model to profile basal mitophagy and macroautophagy in the adult and developing eye. We report that ocular macroautophagy is widespread, but surprisingly mitophagy does not always follow the same pattern of occurrence. We observe low levels of mitophagy in the lens and ciliary body, in stark contrast to the high levels of general MAP1LC3-dependent macroautophagy in these regions. We uncover a striking reversal of this process in the adult retina, where mitophagy accounts for a larger degree of the macroautophagy taking place, specifically in the photoreceptor neurons of the outer nuclear layer. We also show the developmental regulation of autophagy in a variety of ocular tissues. In particular, mitophagy in the adult mouse retina is reversed in localization during the latter stages of development. Our work thus defines the landscape of mitochondrial homeostasis in the mammalian eye, and in doing so highlights the selective nature of autophagy in vivo and the specificity of the reporters used. Abbreviations: ATG: autophagy related; GFP: green fluorescent protein; LC3: microtubule associated protein 1 light chain 3; ONH: optic nerve head; ONL: outer nuclear layer; RPE: retinal pigment epithelium.
Collapse
Affiliation(s)
- Thomas G. McWilliams
- MRC Protein Phosphorylation and Ubiquitylation Unit, Sir James Black Centre School of Life Sciences, University of Dundee, Dundee, UK
- Translational Stem Cell Biology & Metabolism Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, Finland
| | - Alan R. Prescott
- Dundee Imaging Facility, School of Life Sciences, University of Dundee, Dundee, UK
| | - Beatriz Villarejo-Zori
- Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas, CSIC, Madrid, Spain
| | - Graeme Ball
- Dundee Imaging Facility, School of Life Sciences, University of Dundee, Dundee, UK
| | - Patricia Boya
- Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas, CSIC, Madrid, Spain
| | - Ian G. Ganley
- MRC Protein Phosphorylation and Ubiquitylation Unit, Sir James Black Centre School of Life Sciences, University of Dundee, Dundee, UK
| |
Collapse
|
34
|
Ocular biopharmaceutics: impact of modeling and simulation on topical ophthalmic formulation development. Drug Discov Today 2019; 24:1587-1597. [PMID: 30959112 DOI: 10.1016/j.drudis.2019.04.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Revised: 03/07/2019] [Accepted: 04/02/2019] [Indexed: 12/12/2022]
Abstract
The estimation of ocular pharmacokinetics (PK) in various eye tissues is limited because of sampling challenges. Computational modeling and simulation (M&S) tools underpinning the elucidation of drug access routes and prediction of ocular exposure are essential for the mechanistic assessment of biopharmaceutics in the eye. Therefore, theoretical and experimental evaluation of ocular absorption and transit models is necessary. Biopharmaceutical parameter sensitivity analysis based on permeability and drug dose illustrates utility in ocular drug delivery assessment, which could have innovative and cost-saving impacts on ophthalmic product development and therapeutic bioequivalence (BE) evaluations.
Collapse
|
35
|
Coman IC, Al Hammoud M, Tudosescu R, Iancu R, Barac C, Popa CA. The effects of prostaglandins and endocannabinoids on iris arterial vascularization in Wistar rats - Experimental analysis. Rom J Ophthalmol 2019; 63:135-141. [PMID: 31334391 PMCID: PMC6626926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/25/2019] [Indexed: 12/01/2022] Open
Abstract
Introduction: The iris vascular supply originates in the anterior and long posterior ciliary arteries. The endothelium influences local blood flow by releasing endothelium relaxing and contracting substances. From a functional perspective, the ocular vascular tonus adjustment is humoral and neural dependent. Objectives: The present article aims to evaluate the possible implications of topical administration of selective COX2 and nonselective COX inhibitors generically named nonsteroidal anti-inflammatory drugs (NSAIDs) and their possible interactions with the endocannabinoid system and the way they could interfere with the vascular tone at the level of ocular iris territory in Wistar rats. Materials and methods: Experimental protocol on Wistar rats was performed in accordance with present laws regarding animal welfare and ethics in animal experiments (Directive 86/ 609EEC/ 1986; Romanian Law 205/ 2004; Romanian Laws 206/ 2004, 471/ 2002 and 9/ 2008; Romanian Order 143/ 400). The studied substances were instilled topically under general anesthesia, and images of the rat iris vessels were captured over a period of 10 minutes. The obtained images were further analyzed using an appropriate hardware and software program. Results: The nonselective NSAIDs induced vascular dilation in the iris vessels, while the selective COX2 inhibitors determined a variable degree of vasoconstriction. Conclusion: In view of the results of this experiment and the added evidence found in literature, we consider that further research will show the potential benefits for the additional use of NSAIDs in ocular pathology, otherwise unaffected by this medication until the present time (for example, glaucoma treatment).
Collapse
Affiliation(s)
- Ioana-Cristina Coman
- Ophthalmology Department, University Emergency Hospital, Bucharest, Romania
- Faculty of Medicine, ”Carol Davila” University of Medicine and Pharmacy, Bucharest, Romania
| | | | - Ruxandra Tudosescu
- Ophthalmology Department, University Emergency Hospital, Bucharest, Romania
- Ophthalmology Department, Regina Maria Private Clinic, Bucharest, Romania
| | - Raluca Iancu
- Ophthalmology Department, University Emergency Hospital, Bucharest, Romania
- Faculty of Medicine, ”Carol Davila” University of Medicine and Pharmacy, Bucharest, Romania
| | - Cosmina Barac
- Ophthalmology Department, Braila Emergency Hospital, Romania
| | - Cherecheanu Alina Popa
- Ophthalmology Department, University Emergency Hospital, Bucharest, Romania
- Faculty of Medicine, ”Carol Davila” University of Medicine and Pharmacy, Bucharest, Romania
| |
Collapse
|
36
|
Systemic Hypertension Effects on the Ciliary Body and Iris. An Immunofluorescence Study with Aquaporin 1, Aquaporin 4, and Na⁺, K⁺ ATPase in Hypertensive Rats. Cells 2018; 7:cells7110210. [PMID: 30428541 PMCID: PMC6262422 DOI: 10.3390/cells7110210] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 11/08/2018] [Accepted: 11/09/2018] [Indexed: 11/16/2022] Open
Abstract
Aquaporin 1 (AQP1) and aquaporin 4 (AQP4) have been identified in the eye as playing an essential role in the formation of the aqueous humor along with the Na+/K+ ATPase pump. Different authors have described the relationship between blood pressure, aqueous humor production, and intraocular pressure with different conclusions, with some authors supporting a positive correlation between blood pressure and intraocular pressure while others disagree. The aim of this work was to study the effect of high blood pressure on the proteins involved in the production of aqueous humor in the ciliary body (CB) and iris. For this purpose, we used the eyes of spontaneously hypertensive rats (SHR) and their control Wistar-Kyoto rats (WKY). Immunofluorescence was performed in different eye structures to analyze the effects of hypertension in the expression of AQP1, AQP4, and the Na+/K+ ATPase α1 and α2 subunits. The results showed an increase in AQP1 and Na+/K+ ATPase α1 and a decrease in AQP4 and Na+/K+ ATPase α2 in the CB of SHR, while an increase in AQP4 and no significant differences in AQP1 were found in the iris. Therefore, systemic hypertension produced changes in the proteins implicated in the movement of water in the CB and iris that could influence the production rate of aqueous humor, which would be affected depending on the duration of systemic hypertension.
Collapse
|
37
|
Gerhart J, Withers C, Gerhart C, Werner L, Mamalis N, Bravo-Nuevo A, Scheinfeld V, FitzGerald P, Getts R, George-Weinstein M. Myo/Nog cells are present in the ciliary processes, on the zonule of Zinn and posterior capsule of the lens following cataract surgery. Exp Eye Res 2018; 171:101-105. [PMID: 29559302 PMCID: PMC6085112 DOI: 10.1016/j.exer.2018.03.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 03/12/2018] [Accepted: 03/16/2018] [Indexed: 12/11/2022]
Abstract
Myo/Nog cells, named for their expression of MyoD and noggin, enter the eye during early stages of embryonic development. Their release of noggin is critical for normal morphogenesis of the lens and retina. Myo/Nog cells are also present in adult eyes. Single nucleated skeletal muscle cells designated as myofibroblasts arise from Myo/Nog cells in cultures of lens tissue. In this report we document the presence of Myo/Nog cells in the lens, ciliary body and on the zonule of Zinn in mice, rabbits and humans. Myo/Nog cells were rare in all three structures. Their prevalence increased in the lens and ciliary body of rabbits 24 h following cataract surgery. Rabbits developed posterior capsule opacification (PCO) within one month of surgery. The number of Myo/Nog cells continued to be elevated in the lens and ciliary body. Myo/Nog cells containing alpha smooth muscle actin and striated muscle myosin were present on the posterior capsule and overlaid deformations in the capsule. Myo/Nog cells also were present on the zonule fibers and external surface of the posterior capsule. These findings suggest that Myo/Nog contribute to PCO and may use the zonule fibers to migrate between the ciliary processes and lens.
Collapse
Affiliation(s)
| | - Colleen Withers
- Philadelphia College of Osteopathic Medicine, Philadelphia, PA, USA
| | - Colby Gerhart
- Philadelphia College of Osteopathic Medicine, Philadelphia, PA, USA
| | - Liliana Werner
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA
| | - Nick Mamalis
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA
| | | | | | - Paul FitzGerald
- Department of Cell Biology and Human Anatomy, School of Medicine, University of California Davis, Davis, CA, USA
| | | | | |
Collapse
|
38
|
Ashok A, Karmakar S, Chandel R, Ravikumar R, Dalal S, Kong Q, Singh N. Prion protein modulates iron transport in the anterior segment: Implications for ocular iron homeostasis and prion transmission. Exp Eye Res 2018; 175:1-13. [PMID: 29859760 PMCID: PMC6167182 DOI: 10.1016/j.exer.2018.05.031] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 05/17/2018] [Accepted: 05/30/2018] [Indexed: 12/14/2022]
Abstract
Iron is an essential biometal in the aqueous humor, the principal source of nutrients for the avascular cornea and the lens. Here, we explored whether the ciliary body (CB), the source of aqueous humor, transports iron, and if the prion protein (PrPC) facilitates this process as in the outer retina. Using a combination of human, bovine, and mouse eyes as models, we report the expression of iron export proteins ferroportin and ceruloplasmin, and major iron uptake and storage proteins transferrin, transferrin receptor, and ferritin in the ciliary epithelium, indicating active exchange of iron at this site. Ferroportin and transferrin receptor are also expressed in the corneal endothelium. However, the relative expression of iron export and uptake proteins suggests export from the ciliary epithelium and import by corneal endothelium. In addition, abundant expression of PrPC, a ferrireductase that facilitates iron transport, is noted in pigmented and non-pigmented epithelium of the CB, posterior pigmented epithelium of the iris, corneal endothelium and epithelium, and lens epithelium. Notably, majority of PrPC in the ciliary epithelium is cleaved at the β-site as in retinal pigment epithelial cells, suggesting a role in iron transport. Most of the PrPC in the cornea, however, is full-length, and susceptible to aggregation by intracerebrally inoculated PrP-scrapie, an infectious conformation of PrPC responsible for human and animal prion disorders. Soluble PrPC is present in the aqueous and vitreous humor, a provocative observation with significant implications. Together, these observations suggest independent cycling of iron in the anterior segment, and a prominent role of PrPC in this process. Aggregation of PrPC in the cornea of PrP-scrapie-infected animals raises the alarming possibility of transmission of animal prions through corneal abrasions.
Collapse
Affiliation(s)
- Ajay Ashok
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Shilpita Karmakar
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Rajeev Chandel
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Ranjana Ravikumar
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Stuti Dalal
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Qingzhong Kong
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Neena Singh
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA.
| |
Collapse
|
39
|
Rimpelä AK, Reinisalo M, Hellinen L, Grazhdankin E, Kidron H, Urtti A, del Amo EM. Implications of melanin binding in ocular drug delivery. Adv Drug Deliv Rev 2018; 126:23-43. [PMID: 29247767 DOI: 10.1016/j.addr.2017.12.008] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Revised: 10/04/2017] [Accepted: 12/11/2017] [Indexed: 12/20/2022]
Abstract
Pigmented ocular tissues contain melanin within the intracellular melanosomes. Drugs bind to melanin at varying extent that ranges from no binding to extensive binding. Binding may lead to drug accumulation to the pigmented tissues and prolonged drug retention in the melanin containing cells. Therefore, melanin binding is an important feature that affects ocular drug delivery and biodistribution, but this topic has not been reviewed since 1998. In this review, we present current knowledge on ocular melanin, melanosomes and binding of drugs to pigmented cells and tissues. In vitro, in vivo and in silico methods in the field were critically evaluated, because the literature in this field can be confusing if the reader does not properly understand the methodological aspects. Literature analysis includes a comprehensive table of literature data on melanin binding of drugs. Furthermore, we aimed to give some insights beyond the current literature by making a chemical structure based classification model for melanin binding of drugs and kinetic simulations that revealed significant interplay between melanin binding and drug permeability across the melanosomal and plasma membranes. Overall, more mechanistic and systematic research is needed before the impact of melanin binding on ocular drug delivery can be properly understood and predicted.
Collapse
|
40
|
Sallam AB, Kirkland KA, Barry R, Soliman MK, Ali TK, Lightman S. A Review of Antimicrobial Therapy for Infectious Uveitis of the Posterior Segment. MEDICAL HYPOTHESIS, DISCOVERY & INNOVATION OPHTHALMOLOGY JOURNAL 2018; 7:140-155. [PMID: 30505865 PMCID: PMC6229674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Treatment of infectious posterior uveitis represents a therapeutic challenge for ophthalmologists. The eye is a privileged site, maintained by blood ocular barriers, which limits penetration of systemic antimicrobials into the posterior segment. In addition, topical and subconjunctival therapies are incapable of producing sufficient drug concentrations, intraocularly. Posterior infectious uveitis can be caused by bacteria, virus, fungi, or protozoa. Mode of treatment varies greatly based on the infectious etiology. Certain drugs have advantages over others in the treatment of infectious uveitis. Topical and systemic therapies are often employed in the treatment of ocular infection, yet the route of treatment can have limitations based on penetration, concentration, and duration. The introduction of intravitreal antimicrobial therapy has advanced the management of intraocular infections. Being able to bypass blood-ocular barriers allows high drug concentrations to be delivered directly to the posterior segment with minimal systemic absorption. However, because the difference between the therapeutic and the toxic doses of some antimicrobial drugs falls within a narrow concentration range, intravitreal therapy could be associated with ocular toxicity risks. In many cases of infectious uveitis, combination of intravitreal and systemic therapies are necessary. In this comprehensive review, the authors aimed at reviewing clinically relevant data regarding intraocular and systemic antimicrobial therapy for posterior segment infectious uveitis. The review also discussed the evolving trends in intraocular treatment, and elaborated on antibiotic pharmacokinetics and pharmacodynamics, efficacy, and adverse effects.
Collapse
Affiliation(s)
- Ahmed B Sallam
- Jones Eye Institute, University of Arkansas for Medical Sciences, Arkansas, USA
| | - Kyle A. Kirkland
- Jones Eye Institute, University of Arkansas for Medical Sciences, Arkansas, USA
| | - Richard Barry
- Department of Ophthalmology, Gloucestershire Hospitals NHS Foundation Trust, Gloucester, UK
| | | | - Tayyeba K Ali
- Jones Eye Institute, University of Arkansas for Medical Sciences, Arkansas, USA
| | - Sue Lightman
- UCL Institute of Ophthalmology, Moorfields Eye Hospital, London, UK
| |
Collapse
|
41
|
Differential volume regulation and calcium signaling in two ciliary body cell types is subserved by TRPV4 channels. Proc Natl Acad Sci U S A 2016; 113:3885-90. [PMID: 27006502 DOI: 10.1073/pnas.1515895113] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Fluid secretion by the ciliary body plays a critical and irreplaceable function in vertebrate vision by providing nutritive support to the cornea and lens, and by maintaining intraocular pressure. Here, we identify TRPV4 (transient receptor potential vanilloid isoform 4) channels as key osmosensors in nonpigmented epithelial (NPE) cells of the mouse ciliary body. Hypotonic swelling and the selective agonist GSK1016790A (EC50 ∼33 nM) induced sustained transmembrane cation currents and cytosolic [Formula: see text] elevations in dissociated and intact NPE cells. Swelling had no effect on [Formula: see text] levels in pigment epithelial (PE) cells, whereas depolarization evoked [Formula: see text] elevations in both NPE and PE cells. Swelling-evoked [Formula: see text] signals were inhibited by the TRPV4 antagonist HC067047 (IC50 ∼0.9 μM) and were absent in Trpv4(-/-) NPE. In NPE, but not PE, swelling-induced [Formula: see text] signals required phospholipase A2 activation. TRPV4 localization to NPE was confirmed with immunolocalization and excitation mapping approaches, whereas in vivo MRI analysis confirmed TRPV4-mediated signals in the intact mouse ciliary body. Trpv2 and Trpv4 were the most abundant vanilloid transcripts in CB. Overall, our results support a model whereby TRPV4 differentially regulates cell volume, lipid, and calcium signals in NPE and PE cell types and therefore represents a potential target for antiglaucoma medications.
Collapse
|
42
|
Expression analysis of ADAM17 during mouse eye development. Ann Anat 2011; 194:334-8. [PMID: 22055939 DOI: 10.1016/j.aanat.2011.10.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2011] [Revised: 09/26/2011] [Accepted: 10/07/2011] [Indexed: 01/11/2023]
Abstract
ADAM17 (a disintegrin and metallopeptidase domain 17) is crucial for eye morphogenesis. In this study we analysed the expression pattern of ADAM17 during mouse eye development. ADAM17 expression in adult retina was examined using the reverse transcription-polymerase chain reaction (RT-PCR) and verification of the RT-PCR products by DNA sequencing. Immunohistochemistry was performed to evaluate the ADAM17 expression pattern in mouse eyes at developmental stages of embryonic day (E) 12, E14, E16, E18, postnatal day (P) 0, P1, P4, P7, P14, P 30 and P175 (adult). We detected ADAM17 mRNA in adult retina tissue. ADAM17 protein was expressed in non-pigmented ciliary epithelial cells and in retinal vessels from P7 onwards during eye development. In corneal epithelial cells and endothelium, ADAM17 protein was present from P14 onwards. Although, mice in which the functional ADAM17 gene is significantly reduced develop multiple eye malformations, the expression of ADAM17 is not ubiquitous over the entire eye. Its expression pattern during development suggests that not only TNF-alpha but additional membrane-anchored substrates of ADAM17 play an important role in eye formation.
Collapse
|
43
|
Toker M, Bagcivan I, Topalkara A, Karadas B, Durmus N, Parlak A, Kaya T. Nifedipine enhances the relaxant effects of cyclo-oxygenase inhibitors on the bovine ciliary muscle. J Ocul Pharmacol Ther 2008; 24:537-41. [PMID: 19046136 PMCID: PMC2958440 DOI: 10.1089/jop.2007.0129] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2007] [Accepted: 07/02/2008] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND/AIMS The inhibition of cyclo-oxygenase (COX) enzymes and the blockade of Ca (2+) channels play an important role in the regulation of smooth muscle relaxation. This study was designed to investigate the relaxant effects of celecoxib, DFU (5,5-dimethyl-3-(3-fluorophenyl)-4-(4-methylsulphonyl)phenyl-2(5H)-furanone), and indomethacin, cyclo-oxygenase (COX-1 and -2) inhibitors, in the absence or presence of a nifedipine, L-type Ca(2+) channel blocker, on bovine ciliary muscle. METHODS Ciliary muscle strips (n = 12) were mounted in organ baths and tested for changes in isometric tension in response to celecoxib, DFU, and indomethacin. The relaxant effects of celecoxib, DFU, and indomethacin on carbachol-induced contractions in the presence or absence of nifedipine were investigated. RESULTS Celecoxib (10(-7)-10(-4) M), DFU (10(-7)-10(-4) M), indomethacin (10(-7)-10(-4) M), and nifedipine (10(-7)-10(-4) M) inhibited the carbachol-induced contractions in a concentration-dependent manner. The E(max) value of indomethacin was significantly higher than the E(max) values of celecoxib and DFU in ciliary muscle (P < 0.05), with no significant change in pD(2) values (P > 0.05). The relaxation responses by celecoxib, DFU, and indomethacin were significantly increased in the presence of nifedipine (10(-6) M). There were no significant differences between pEC50 and values of celecoxib, DFU, and indomethacin in the absence of nifedipine (10(-6) M) (P > 0.05), but E(max)values were significantly increased (P < 0.05). CONCLUSIONS These results suggest that the celecoxib, DFU, and indomethacin cause relaxation in ciliary muscle precontracted with carbachol. Blockade of calcium channels with nifedipine in ciliary muscle may increase the relaxant effect of celecoxib, DFU, and indomethacin. The topical or systemic use of celecoxib, DFU, and indomethacin with nifedipine can cause blurred near vision due to ciliary muscle relaxation, and in ocular pain conditions caused by ciliary spasm, the pain can be decreased more easily by combined use of these drugs.
Collapse
Affiliation(s)
- Mustafa Toker
- Department of Ophthalmology, Cumhuriyet University School of Medicine, Sivas, Turkey.
| | | | | | | | | | | | | |
Collapse
|
44
|
Topalkara A, Karadas B, Toker MI, Kaya T, Durmus N, Turgut B. Relaxant effects of β-adrenoceptor agonist formoterol and BRL 37344 on bovine iris sphincter and ciliary muscle. Eur J Pharmacol 2006; 548:144-9. [PMID: 16973159 DOI: 10.1016/j.ejphar.2006.08.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2006] [Revised: 07/22/2006] [Accepted: 08/02/2006] [Indexed: 11/29/2022]
Abstract
We investigated the relaxant effect of beta2-adrenoceptor agonist formoterol and beta3-adrenoceptor agonist BRL 37344 on bovine iris sphincter and ciliary muscle and measured cAMP and cGMP levels. Iris sphincter (n = 16) and ciliary muscle (n = 16) strips were mounted in organ baths and tested for changes in isometric tension in response to formoterol and BRL 37344. Their relaxant effects on serotonin-induced contractions in the presence or absence of metoprolol, ICI 118.551 and SR 59230A (beta1-, beta2-, beta3-adrenoceptor antagonist, respectively) were investigated. Their effects on cAMP and cGMP levels in iris sphincter (n = 12) and ciliary muscle (n = 12) were evaluated. Formoterol (10(-11)-10(-5) M) and BRL 37344 (10(-10)-10(-5) M) decreased the serotonine-induced contractions in a concentration-dependent manner. Emax values of formoterol were significantly higher than those of BRL 37344 in iris sphincter and ciliary muscle, with no significant change in pD2 values. The relaxation responses by formoterol and BRL 37344 were antagonized with ICI 118.551 (10(-6) M) and SR 59230A (10(-6) M). cAMP levels of formoterol- and BRL 37344-treated tissues were significantly higher than those of the control tissues. cGMP levels of BRL 37344-treated tissues were significantly higher than those of control tissues, but this effect of BRL 37344 was less significant than its effect on cAMP levels. beta-adrenoceptor relaxation responses in bovine iris sphincter and ciliary muscle are mediated by a mixed population of beta2- and beta3-adrenoceptor subtypes, with a predominant contribution of cAMP. Potency of formoterol and BRL 37344 was similar, but efficacy of formoterol was better than BRL 37344.
Collapse
Affiliation(s)
- Aysen Topalkara
- Department of Ophthalmology, Cumhuriyet University School of Medicine, 58140 Sivas, Turkey.
| | | | | | | | | | | |
Collapse
|