1
|
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
|
2
|
Becker S, L'Ecuyer Z, Jones BW, Zouache MA, McDonnell FS, Vinberg F. Modeling complex age-related eye disease. Prog Retin Eye Res 2024; 100:101247. [PMID: 38365085 PMCID: PMC11268458 DOI: 10.1016/j.preteyeres.2024.101247] [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: 08/15/2023] [Revised: 01/31/2024] [Accepted: 02/02/2024] [Indexed: 02/18/2024]
Abstract
Modeling complex eye diseases like age-related macular degeneration (AMD) and glaucoma poses significant challenges, since these conditions depend highly on age-related changes that occur over several decades, with many contributing factors remaining unknown. Although both diseases exhibit a relatively high heritability of >50%, a large proportion of individuals carrying AMD- or glaucoma-associated genetic risk variants will never develop these diseases. Furthermore, several environmental and lifestyle factors contribute to and modulate the pathogenesis and progression of AMD and glaucoma. Several strategies replicate the impact of genetic risk variants, pathobiological pathways and environmental and lifestyle factors in AMD and glaucoma in mice and other species. In this review we will primarily discuss the most commonly available mouse models, which have and will likely continue to improve our understanding of the pathobiology of age-related eye diseases. Uncertainties persist whether small animal models can truly recapitulate disease progression and vision loss in patients, raising doubts regarding their usefulness when testing novel gene or drug therapies. We will elaborate on concerns that relate to shorter lifespan, body size and allometries, lack of macula and a true lamina cribrosa, as well as absence and sequence disparities of certain genes and differences in their chromosomal location in mice. Since biological, rather than chronological, age likely predisposes an organism for both glaucoma and AMD, more rapidly aging organisms like small rodents may open up possibilities that will make research of these diseases more timely and financially feasible. On the other hand, due to the above-mentioned anatomical and physiological features, as well as pharmacokinetic and -dynamic differences small animal models are not ideal to study the natural progression of vision loss or the efficacy and safety of novel therapies. In this context, we will also discuss the advantages and pitfalls of alternative models that include larger species, such as non-human primates and rabbits, patient-derived retinal organoids, and human organ donor eyes.
Collapse
Affiliation(s)
- Silke Becker
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA
| | - Zia L'Ecuyer
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA
| | - Bryan W Jones
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA
| | - Moussa A Zouache
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA
| | - Fiona S McDonnell
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA; Biomedical Engineering, University of Utah, Salt Lake City, UT, USA
| | - Frans Vinberg
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA; Biomedical Engineering, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
3
|
Agarwal R, Iezhitsa I. Genetic rodent models of glaucoma in representing disease phenotype and insights into the pathogenesis. Mol Aspects Med 2023; 94:101228. [PMID: 38016252 DOI: 10.1016/j.mam.2023.101228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 10/31/2023] [Accepted: 11/11/2023] [Indexed: 11/30/2023]
Abstract
Genetic rodent models are widely used in glaucoma related research. With vast amount of information revealed by human studies about genetic correlations with glaucoma, use of these models is relevant and required. In this review, we discuss the glaucoma endophenotypes and importance of their representation in an experimental animal model. Mice and rats are the most popular animal species used as genetic models due to ease of genetic manipulations in these animal species as well as the availability of their genomic information. With technological advances, induction of glaucoma related genetic mutations commonly observed in human is possible to achieve in rodents in a desirable manner. This approach helps to study the pathobiology of the disease process with the background of genetic abnormalities, reveals potential therapeutic targets and gives an opportunity to test newer therapeutic options. Various genetic manipulation leading to appearance of human relevant endophenotypes in rodents indicate their relevance in glaucoma pathology and the utility of these rodent models for exploring various aspects of the disease related to targeted mutation. The molecular pathways involved in the pathophysiology of glaucoma leading to elevated intraocular pressure and the disease hallmark, apoptosis of retinal ganglion cells and optic nerve degeneration, have been extensively explored in genetic rodent models. In this review, we discuss the consequences of various genetic manipulations based on the primary site of pathology in the anterior or the posterior segment. We discuss how these genetic manipulations produce features in rodents that can be considered a close representation of disease phenotype in human. We also highlight several molecular mechanisms revealed by using genetic rodent models of glaucoma including those involved in increased aqueous outflow resistance, loss of retinal ganglion cells and optic neuropathy. Lastly, we discuss the limitations of the use of genetic rodent models in glaucoma related research.
Collapse
Affiliation(s)
- Renu Agarwal
- School of Medicine, International Medical University, Malaysia.
| | - Igor Iezhitsa
- School of Medicine, International Medical University, Malaysia
| |
Collapse
|
4
|
Li HL, Ren R, Gong H. Segmental Unconventional Outflow in Mouse Eyes. Invest Ophthalmol Vis Sci 2023; 64:26. [PMID: 38117243 PMCID: PMC10741088 DOI: 10.1167/iovs.64.15.26] [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: 09/09/2023] [Accepted: 11/28/2023] [Indexed: 12/21/2023] Open
Abstract
Purpose To investigate the flow pattern in unconventional outflow and its correlation with conventional outflow in mouse eyes. Methods Fluorescent microspheres were injected into the anterior chamber of one eye of anesthetized C57BL/6J mice (n = 4), followed by perfused fixation with 4% paraformaldehyde in situ after 45 minutes. Post-euthanasia, the injected eyes were enucleated, further immersion fixed, and dissected into 12 equal radial segments. Both sides of each segment were imaged using a confocal microscope after nuclear counterstaining. Both unconventional and conventional outflow patterns of each eye were analyzed by ImageJ and ZEN 2.3 imaging software. Results Segmental outflow patterns were observed in both the ciliary body (CB) and the supraciliary space and suprachoroidal space (SCS). In the CB, the tracer intensity was the lowest at 12 o'clock and highest at 9 o'clock, whereas in the SCS it was the lowest at 2 o'clock and the highest at 10 o'clock. Consequently, a segmental unconventional outflow was observed, with the lowest and highest flow regions in the superior and temporal quadrants, respectively. The overall segmental uveoscleral outflow has no correlation with trabecular outflow (P > 0.05). Four different outflow patterns were observed: (1) low-flow regions in both outflows, (2) primarily a high-flow region in conventional outflow, (3) primarily a high-flow region in unconventional outflow, and (4) high-flow regions in both outflows. Conclusions Uveoscleral outflow is segmental and unrelated to the trabecular segmental outflow. These findings will lead to future studies to identify the best location for the placement of drainage devices and drug delivery.
Collapse
Affiliation(s)
- Hoi-Lam Li
- Ophthalmology, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, United States
| | - Ruiyi Ren
- Ophthalmology, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, United States
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Zhejiang, China
| | - Haiyan Gong
- Ophthalmology, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, United States
- Anatomy and Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, United States
| |
Collapse
|
5
|
Strohmaier CA, Wanderer D, Zhang X, Agarwal D, Toomey CB, Wahlin K, Zhang HF, Stamer WD, Weinreb RN, McDonnell FS, Huang AS. Greater Outflow Facility Increase After Targeted Trabecular Bypass in Angiographically Determined Low-low Regions. Ophthalmol Glaucoma 2023; 6:570-579. [PMID: 37348815 PMCID: PMC10917462 DOI: 10.1016/j.ogla.2023.06.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 05/31/2023] [Accepted: 06/14/2023] [Indexed: 06/24/2023]
Abstract
PURPOSE To investigate the impact of trabecular bypass surgery targeted to angiographically determined high- vs. low-aqueous humor outflow areas on outflow facility (C) and intraocular pressure (IOP). DESIGN Ex vivo comparative study. SUBJECTS Postmortem ex vivo porcine and human eyes. METHODS Porcine (n = 14) and human (n = 13) whole globes were acquired. In both species, anterior segments were dissected, mounted onto a perfusion chamber, and perfused using Dulbecco's phosphate buffered solution containing glucose in a constant flow paradigm to achieve a stable baseline. Fluorescein was perfused into the anterior chamber and used to identify baseline segmental high- and low-flow regions of the conventional outflow pathways. The anterior segments were divided into 2 groups, and a 5 mm needle goniotomy was performed in either a high- or low-flow area. Subsequently, C and IOP were quantitatively reassessed and compared between surgery in baseline "high-flow" and "low-flow" region eyes followed by indocyanine green angiography. MAIN OUTCOME MEASURES Outflow facility. RESULTS In all eyes, high- and low-flow segments could be identified. Performing a 5-mm goniotomy increased outflow facility to a variable extent depending on baseline flow status. In the porcine high-flow group, C increased from 0.31 ± 0.09 to 0.39 ± 0.09 μL/mmHg/min (P = 0.12). In the porcine low-flow group, C increased from 0.29 ± 0.03 to 0.56 ± 0.10 μL/mmHg/min (P < 0.001). In the human high-flow group, C increased from 0.38 ± 0.20 to 0.41 ± 0.20 μL/mmHg/min (P = 0.02). In the human low-flow group, C increased from 0.25 ± 0.11 to 0.32 ± 0.11 μL/mmHg/min (<0.001). There was statistically significant greater increase in C for eyes where surgery was targeted to baseline low-flow regions in both porcine (0.07 ± 0.09 vs. 0.27 ± 0.13, P = 0.007 μL/mmHg/min, high vs low flow) and human eyes (0.03 ± 0.03 vs. 0.07 ± 0.02, P = 0.03 μL/mmHg/min, high vs. low flow). CONCLUSIONS Targeting surgery to low-flow areas of the trabecular meshwork yields higher overall facility increase and IOP reduction compared to surgery in high-flow areas. FINANCIAL DISCLOSURE(S) Proprietary or commercial disclosure may be found in the Footnotes and Disclosures at the end of this article.
Collapse
Affiliation(s)
- Clemens A Strohmaier
- Department of Ophthalmology and Optometry, Kepler University Hospital, Johannes Kepler University, Linz, Austria; The Viterbi Family Department of Ophthalmology, Hamilton Glaucoma Center, Shiley Eye Institute, University of California, San Diego, California.
| | - Daniel Wanderer
- The Viterbi Family Department of Ophthalmology, Hamilton Glaucoma Center, Shiley Eye Institute, University of California, San Diego, California
| | - Xiaowei Zhang
- The Viterbi Family Department of Ophthalmology, Hamilton Glaucoma Center, Shiley Eye Institute, University of California, San Diego, California
| | - Devansh Agarwal
- The Viterbi Family Department of Ophthalmology, Hamilton Glaucoma Center, Shiley Eye Institute, University of California, San Diego, California
| | - Christopher B Toomey
- The Viterbi Family Department of Ophthalmology, Hamilton Glaucoma Center, Shiley Eye Institute, University of California, San Diego, California
| | - Karl Wahlin
- The Viterbi Family Department of Ophthalmology, Hamilton Glaucoma Center, Shiley Eye Institute, University of California, San Diego, California
| | - Hao F Zhang
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois
| | - W Daniel Stamer
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
| | - Robert N Weinreb
- The Viterbi Family Department of Ophthalmology, Hamilton Glaucoma Center, Shiley Eye Institute, University of California, San Diego, California
| | | | - Alex S Huang
- The Viterbi Family Department of Ophthalmology, Hamilton Glaucoma Center, Shiley Eye Institute, University of California, San Diego, California
| |
Collapse
|
6
|
Kicińska AK, Rękas M. Alternative application of an iTrack microcatheter and canaloplasty: case report and literature review. Expert Opin Drug Deliv 2023; 20:1201-1208. [PMID: 37700455 DOI: 10.1080/17425247.2023.2256657] [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: 06/19/2023] [Accepted: 09/05/2023] [Indexed: 09/14/2023]
Abstract
INTRODUCTION Glaucoma is the leading cause of irreversible blindness worldwide. Schlemm's canal surgery using an iTrack flexible microcatheter has become popular because of its high quality-of-life issues and the growing demand for less invasive but effective procedures. The unique design of the microcatheter makes it a multimodal tool, which can be used not only in the field of antiglaucoma surgery but also as a drug delivery system to treat various conditions. AREAS COVERED This review presents an update on the selected aspects of a drug delivery system using the iTrack microcatheter, including glaucoma gene therapy and posterior-segment diseases, both in animal models and human patients. The authors also report the case of a patient with branch retinal vein occlusion treated with suprachoroidal bevacizumab in the submacular region administered with the iTrack catheter. EXPERT OPINION The findings presented in this study may indicate that the application of a microcatheter in open-angle glaucoma gene therapy is reasonable and can be combined with full or partial surgical canaloplasty procedures. Translation of this potential into a treatment modality would require overcoming multiple barriers.
Collapse
Affiliation(s)
- Aleksandra K Kicińska
- Department of Ophthalmology, Military Institute of Medicine - National Research Institute, Warsaw, Poland
| | - Marek Rękas
- Department of Ophthalmology, Military Institute of Medicine - National Research Institute, Warsaw, Poland
| |
Collapse
|
7
|
Ren R, Humphrey AA, Kopczynski C, Gong H. Rho Kinase Inhibitor AR-12286 Reverses Steroid-Induced Changes in Intraocular Pressure, Effective Filtration Areas, and Morphology in Mouse Eyes. Invest Ophthalmol Vis Sci 2023; 64:7. [PMID: 36734964 PMCID: PMC9907372 DOI: 10.1167/iovs.64.2.7] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 01/11/2023] [Indexed: 02/04/2023] Open
Abstract
Purpose We investigated mechanisms of reduction of intraocular pressure (IOP) by Rho kinase inhibitor AR-12286 in steroid-induced ocular hypertension (SIOH). Methods C57BL/6 mice (N = 56) were randomly divided into Saline, dexamethasone (DEX), DEX + AR-12286, and DEX-discontinuation (DEX-DC) groups. IOP was measured weekly during the first four weeks in all groups. Beginning at week 5, the DEX-DC group was followed without treatment until IOP returned to normal, and the other groups were treated as assigned with IOP measured every other day for another week. Fluorescent tracer was injected into the anterior chamber to visualize the outflow pattern in the trabecular meshwork (TM) and TM effective filtration area (EFA) was determined. Radial sections from both high- and low-tracer regions were processed for electron microscopy. Results AR-12286 reduced IOP in SIOH mouse eyes in one day (P < 0.01). At the end of week 5, mean IOP in the DEX + AR-12286 group was ∼4 mm Hg lower than DEX group (P < 0.001) and ∼2 mm Hg lower than DEX-DC group (P < 0.05). After one-week AR-12286 treatment (P < 0.05) or five-week DC of DEX (P < 0.01), DEX-induced reduction of EFA was rescued and DEX-induced morphological changes in the TM were partially reversed. Conclusions AR-12286 reversed steroid-induced morphological changes in the TM and reduced EFA, which correlated with reduced IOP in SIOH eyes. AR-12286 reduced IOP elevation in SIOH eyes more effectively than discontinuing DEX treatment even when accompanied by continuous DEX treatment. Therefore Rho kinase inhibitors may lower SIOH in patients who rely on steroid treatment.
Collapse
Affiliation(s)
- Ruiyi Ren
- Boston University School of Medicine, Department of Ophthalmology, Boston, Massachusetts, United States
| | - Anne A. Humphrey
- Boston University School of Medicine, Department of Ophthalmology, Boston, Massachusetts, United States
| | - Casey Kopczynski
- Aerie Pharmaceuticals, Inc., Durham, North Carolina, United States
| | - Haiyan Gong
- Boston University School of Medicine, Department of Ophthalmology, Boston, Massachusetts, United States
| |
Collapse
|
8
|
Yu PK, Tay E, An D, Cringle SJ, Morgan WH, Yu DY. Topographic distribution and phenotypic heterogeneity of Schlemm's canal endothelium in human donor eyes. Exp Eye Res 2023; 226:109309. [PMID: 36400284 DOI: 10.1016/j.exer.2022.109309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 11/06/2022] [Accepted: 11/08/2022] [Indexed: 11/17/2022]
Abstract
Endothelium phenotype is known to be closely associated with flow shear stress. This study is to determine the topographic distribution of endothelial cells and the phenotype of different quadrants and regions of Schlemm's canal using human donor eyes. This study infers differences in flow dynamics based on cell shape and intracellular structure. The Schlemm's canal from 15 human donor eyes were either perfusion labelled using silver stain or dissected for float labeling with Phalloidin to enable visualization of endothelial cell border and intracellular structure. Data were acquired for endothelial cells from the outer and inner wall of Schlemm's canal and grouped according to quadrant of origin. Measurements included endothelial cell length, width, area, and aspect ratio and compared between quadrants. Endothelial cells are mostly spindle-shape and the cell size on the outer wall are larger and longer than those from the inner wall. Significant differences in endothelial cell size and shape were seen in different quadrants. The endothelial cells have varied shapes and orientations close to large ostia in the outer wall and remarkably long endothelial cells were found in the walls of collector channels. F-actin aggregation was found at all endothelial cell borders, and inside some of the endothelial cytoplasm. The presence of various spindle shapes, significant phenotype heterogeneity and F-actin aggregation of endothelial cells indicates aqueous humor flow likely creates variations in shear stress within Schlemm's canal. Further investigation of the relationship between the phenotype heterogeneity and hydrodynamics of aqueous flow may help us understand the mechanisms of outflow resistance changes in glaucoma.
Collapse
Affiliation(s)
- Paula K Yu
- Centre for Ophthalmology and Visual Science, The University of Western Australia, Perth, Australia; Lions Eye Institute, The University of Western Australia, Perth, Australia
| | | | - Dong An
- Centre for Ophthalmology and Visual Science, The University of Western Australia, Perth, Australia; Lions Eye Institute, The University of Western Australia, Perth, Australia
| | - Stephen J Cringle
- Centre for Ophthalmology and Visual Science, The University of Western Australia, Perth, Australia; Lions Eye Institute, The University of Western Australia, Perth, Australia
| | - William H Morgan
- Centre for Ophthalmology and Visual Science, The University of Western Australia, Perth, Australia; Lions Eye Institute, The University of Western Australia, Perth, Australia
| | - Dao-Yi Yu
- Centre for Ophthalmology and Visual Science, The University of Western Australia, Perth, Australia; Lions Eye Institute, The University of Western Australia, Perth, Australia.
| |
Collapse
|
9
|
Segmental outflow dynamics in the trabecular meshwork of living mice. Exp Eye Res 2022; 225:109285. [PMID: 36273576 DOI: 10.1016/j.exer.2022.109285] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/10/2022] [Accepted: 10/14/2022] [Indexed: 12/29/2022]
Abstract
Aqueous humour does not drain uniformly through the trabecular meshwork (TM), but rather follows non-uniform or "segmental" routes. In this study, we examined whether segmental outflow patterns in the TM change over time in living mice and whether such changes are affected by age. Segmental outflow patterns were labelled by constant-pressure infusion of fluorescent tracer microparticles into the anterior chamber of anesthetised C57BL/6J mice at 3 or 8 months of age. Two different tracer colours were infused at separate time points with an interval of Δt = 0, 2, 7 or 14 days. In a separate experiment, one tracer was infused in vivo while the second tracer was infused ex vivo after 2 days. The spatial relationship between the two tracer patterns was analysed using the Pearson's correlation coefficient, r. In 3-month-old mice, there was a time-dependent decay in r, which was near unity at Δt = 0 and near zero at Δt = 14 days. In 8-month-old mice, r remained elevated for 14 days. Segmental outflow patterns measured in young mice ex vivo were not significantly different from those measured in vivo after accounting for the expected changes over 2 days. Therefore, segmental outflow patterns are not static in the TM but redistribute over time, achieving near complete loss of correlation by 2 weeks in young healthy mice. There is an age-related decline in the rate at which segmental outflow patterns redistribute in the TM. Further research is needed to understand the dynamic factors controlling segmental outflow.
Collapse
|
10
|
Nikhalashree S, George R, Shantha B, Vijaya L, Sulochana KN, Coral K. Anti-glaucoma medications lowered decorin and altered profibrotic proteins in human tenon's fibroblasts. Exp Eye Res 2022; 224:109199. [PMID: 35878659 DOI: 10.1016/j.exer.2022.109199] [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: 11/29/2021] [Revised: 06/19/2022] [Accepted: 07/18/2022] [Indexed: 11/04/2022]
Abstract
Long term exposure to anti-glaucoma medications (AGMs) leads to an increase in extracellular matrix (ECM) accumulation in primary glaucoma patients. This study aims to evaluate the effect of topical AGMs in primary human tenon's fibroblasts (HTFs) and analyze the expression of profibrotic and anti-fibrotic proteins. Primary HTFs were cultured from patients undergoing cataract (control) and trabeculectomy. The different types of AGMs in single/multiple combinations (BB, PG, AA, CAI, CH, combinations of 3- PG + AA + CAI, 4A- BB + PG + AA + CAI, 4B- BB + PG + CAI + CH and 5- BB + PG + AA + CAI + CH) on chronic exposure were tested for cell viability using MTT assay and morphological alterations. Profibrotic proteins mainly SPARC, LOXL2, COL1A1 and anti-fibrotic DCN were analyzed in treated HTFs using q-PCR and ELISA. Sirius red staining and collagen gel contraction (CGC) assay were performed to assess collagen synthesis and the contractility of HTFs, respectively. Except for AA and CH, the other AGMs at a higher concentration were found to decrease the cell viability of HTFs. The morphology of HTFs were altered on exposure to BB, CH and AA; Profibrotic proteins i.e., SPARC, LOXL2 and COL1A1 were significantly increased (p < 0.05) on exposure to a combination of AGMs with TGF-β1, whereas the anti-fibrotic DCN expression was significantly lowered (p < 0.05) in single/multiple AGM exposure. Sirius red staining showed increased collagen synthesis with combinations of AGMs with TGF-β1. Meanwhile, HTFs showed increased collagen gel contraction with TGF-β1, CAI and CH. This study reveals that altered profibrotic proteins, with significantly lowered DCN on chronic exposure of AGMs in HTFs.
Collapse
Affiliation(s)
- Sampath Nikhalashree
- R.S. Mehta Jain Department of Biochemistry and Cell Biology, KBIRVO Block, Vision Research Foundation, Sankara Nethralaya, Chennai, India; School of Chemical and Biotechnology, SASTRA Deemed-to-be University, Thanjavur, India
| | - Ronnie George
- Smt Jadhavbai Nathmal Singhvee Glaucoma Services, Medical Research Foundation, Sankara Nethralaya, Chennai, India
| | - Balekudaru Shantha
- Smt Jadhavbai Nathmal Singhvee Glaucoma Services, Medical Research Foundation, Sankara Nethralaya, Chennai, India
| | - Lingam Vijaya
- Smt Jadhavbai Nathmal Singhvee Glaucoma Services, Medical Research Foundation, Sankara Nethralaya, Chennai, India
| | | | - Karunakaran Coral
- R.S. Mehta Jain Department of Biochemistry and Cell Biology, KBIRVO Block, Vision Research Foundation, Sankara Nethralaya, Chennai, India.
| |
Collapse
|
11
|
Lewczuk K, Jabłońska J, Konopińska J, Mariak Z, Rękas M. Schlemm's canal: the outflow 'vessel'. Acta Ophthalmol 2022; 100:e881-e890. [PMID: 34519170 PMCID: PMC9293138 DOI: 10.1111/aos.15027] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 06/21/2021] [Accepted: 08/31/2021] [Indexed: 12/26/2022]
Abstract
In a healthy eye, the aqueous humour (AH) flows via the ciliary body and trabecular meshwork into the collector channels, which carry it to the episcleral veins. In glaucoma, a heterogeneous group of eye disorders affecting approximately 60 million individuals worldwide, the juxtacanalicular meshwork offers greater resistance to the outflow of the AH, leading to an increase in outflow resistance that gradually results in elevated intraocular pressure (IOP). The present review comprehensively covers the morphology of Schlemm’s canal (SC) and AH pathways. The path of the AH from the anterior chamber through the trabeculum into suprascleral and conjunctival veins via collector channels is described, and the role of SC in the development of glaucoma and outflow resistance is discussed. Finally, channelography is presented as a precise method of assessing the conventional drainage pathway and facilitating localization of an uncollapsed collector and aqueous veins. Attention is also given to the relationship between aqueous and episcleral veins and heartbeat. Possible directions of future research are proposed.
Collapse
Affiliation(s)
- Katarzyna Lewczuk
- Department of Ophthalmology Military Institute of Medicine Warsaw Poland
| | - Joanna Jabłońska
- Department of Ophthalmology Military Institute of Medicine Warsaw Poland
| | - Joanna Konopińska
- Department of Ophthalmology Medical University in Bialystok Białystok Poland
| | - Zofia Mariak
- Department of Ophthalmology Medical University in Bialystok Białystok Poland
| | - Marek Rękas
- Department of Ophthalmology Military Institute of Medicine Warsaw Poland
| |
Collapse
|
12
|
MacDonald WW, Swaminathan SS, Heo JY, Castillejos A, Hsueh J, Liu BJ, Jo D, Du A, Lee H, Kang MH, Rhee DJ. Effect of SPARC Suppression in Mice, Perfused Human Anterior Segments, and Trabecular Meshwork Cells. Invest Ophthalmol Vis Sci 2022; 63:8. [PMID: 35671048 PMCID: PMC9187959 DOI: 10.1167/iovs.63.6.8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Purpose Secreted protein, acidic and rich in cysteine (SPARC) elevates intraocular pressure (IOP), increases certain structural extracellular matrix (ECM) proteins in the juxtacanalicular trabecular meshwork (JCT), and decreases matrix metalloproteinase (MMP) protein levels in trabecular meshwork (TM) endothelial cells. We investigated SPARC as a potential target for lowering IOP. We hypothesized that suppressing SPARC will decrease IOP, decrease structural JCT ECM proteins, and alter the levels of MMPs and/or their inhibitors. Methods A lentivirus containing short hairpin RNA of human SPARC suppressed SPARC in mouse eyes and perfused cadaveric human anterior segments with subsequent IOP measurements. Immunohistochemistry determined structural correlates. Human TM cell cultures were treated with SPARC suppressing lentivirus. Quantitative reverse transcriptase polymerase chain reaction (PCR), immunoblotting, and zymography determined total RNA, relative protein levels, and MMP enzymatic activity, respectively. Results Suppressing SPARC decreased IOP in mouse eyes and perfused human anterior segments by approximately 20%. Histologically, this correlated to a decrease in collagen I, IV, and VI in both the mouse TM and human JCT regions; in the mouse, fibronectin was also decreased but not in the human. In TM cells, collagen I and IV, fibronectin, MMP-2, and tissue inhibitor of MMP-1 were decreased. Messenger RNA of the aforementioned genes was not changed. Plasminogen activator inhibitor 1 (PAI-1) was upregulated in vitro by quantitative PCR and immunoblotting. MMP-1 activity was reduced in vitro by zymography. Conclusions Suppressing SPARC decreased IOP in mice and perfused cadaveric human anterior segments corresponding to qualitative structural changes in the JCT ECM, which do not appear to be the result of transcription regulation.
Collapse
Affiliation(s)
- William W MacDonald
- Department of Ophthalmology & Visual Sciences, University Hospitals, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States
| | - Swarup S Swaminathan
- Department of Ophthalmology & Visual Sciences, University Hospitals, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States.,Department of Ophthalmology, Bascom Palmer Eye Institute, Miami, Florida, United States
| | - Jae Young Heo
- Department of Ophthalmology & Visual Sciences, University Hospitals, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States
| | - Alexandra Castillejos
- Department of Ophthalmology & Visual Sciences, University Hospitals, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States.,Department of Ophthalmology, Massachusetts Eye & Ear Infirmary, Boston, Massachusetts, United States
| | - Jessica Hsueh
- Department of Ophthalmology & Visual Sciences, University Hospitals, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States
| | - Brian J Liu
- Department of Ophthalmology & Visual Sciences, University Hospitals, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States
| | - Diane Jo
- Department of Ophthalmology & Visual Sciences, University Hospitals, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States
| | - Annie Du
- Department of Ophthalmology & Visual Sciences, University Hospitals, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States
| | - Hyunpil Lee
- Department of Ophthalmology & Visual Sciences, University Hospitals, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States
| | - Min Hyung Kang
- Department of Ophthalmology & Visual Sciences, University Hospitals, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States
| | - Douglas J Rhee
- Department of Ophthalmology & Visual Sciences, University Hospitals, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States
| |
Collapse
|
13
|
McDowell CM, Kizhatil K, Elliott MH, Overby DR, van Batenburg-Sherwood J, Millar JC, Kuehn MH, Zode G, Acott TS, Anderson MG, Bhattacharya SK, Bertrand JA, Borras T, Bovenkamp DE, Cheng L, Danias J, De Ieso ML, Du Y, Faralli JA, Fuchshofer R, Ganapathy PS, Gong H, Herberg S, Hernandez H, Humphries P, John SWM, Kaufman PL, Keller KE, Kelley MJ, Kelly RA, Krizaj D, Kumar A, Leonard BC, Lieberman RL, Liton P, Liu Y, Liu KC, Lopez NN, Mao W, Mavlyutov T, McDonnell F, McLellan GJ, Mzyk P, Nartey A, Pasquale LR, Patel GC, Pattabiraman PP, Peters DM, Raghunathan V, Rao PV, Rayana N, Raychaudhuri U, Reina-Torres E, Ren R, Rhee D, Chowdhury UR, Samples JR, Samples EG, Sharif N, Schuman JS, Sheffield VC, Stevenson CH, Soundararajan A, Subramanian P, Sugali CK, Sun Y, Toris CB, Torrejon KY, Vahabikashi A, Vranka JA, Wang T, Willoughby CE, Xin C, Yun H, Zhang HF, Fautsch MP, Tamm ER, Clark AF, Ethier CR, Stamer WD. Consensus Recommendation for Mouse Models of Ocular Hypertension to Study Aqueous Humor Outflow and Its Mechanisms. Invest Ophthalmol Vis Sci 2022; 63:12. [PMID: 35129590 PMCID: PMC8842499 DOI: 10.1167/iovs.63.2.12] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 12/08/2021] [Indexed: 01/07/2023] Open
Abstract
Due to their similarities in anatomy, physiology, and pharmacology to humans, mice are a valuable model system to study the generation and mechanisms modulating conventional outflow resistance and thus intraocular pressure. In addition, mouse models are critical for understanding the complex nature of conventional outflow homeostasis and dysfunction that results in ocular hypertension. In this review, we describe a set of minimum acceptable standards for developing, characterizing, and utilizing mouse models of open-angle ocular hypertension. We expect that this set of standard practices will increase scientific rigor when using mouse models and will better enable researchers to replicate and build upon previous findings.
Collapse
Affiliation(s)
- Colleen M. McDowell
- Department of Ophthalmology and Visual Sciences, University of Wisconsin–Madison, Madison, Wisconsin, United States
| | | | - Michael H. Elliott
- University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Darryl R. Overby
- Department of Bioengineering, Imperial College London, United Kingdom
| | | | - J. Cameron Millar
- Department of Pharmacology & Neuroscience, and North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Markus H. Kuehn
- Department of Ophthalmology and Visual Sciences and Institute for Vision Research, The University of Iowa; Center for the Prevention and Treatment of Visual Loss, Veterans Affairs Medical Center, Iowa City, Iowa, United States
| | - Gulab Zode
- Department of Pharmacology & Neuroscience, and North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Ted S. Acott
- Ophthalmology and Biochemistry and Molecular Biology, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - Michael G. Anderson
- Department of Molecular Physiology and Biophysics and Department of Ophthalmology and Visual Sciences, The University of Iowa; Center for the Prevention and Treatment of Visual Loss, Veterans Affairs Medical Center, Iowa City, Iowa, United States
| | | | - Jacques A. Bertrand
- Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Terete Borras
- University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | | | - Lin Cheng
- Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, Iowa, United States
| | - John Danias
- SUNY Downstate Health Sciences University, Brooklyn, New York, United States
| | - Michael Lucio De Ieso
- Department of Ophthalmology, Duke Eye Center, Duke University, Durham, North Carolina, United States
| | - Yiqin Du
- Department of Ophthalmology, University of Pittsburgh, Pennsylvania, United States
| | - Jennifer A. Faralli
- Department of Pathology and Laboratory Medicine, University of Wisconsin–Madison, Madison, Wisconsin, United States
| | - Rudolf Fuchshofer
- Institute of Human Anatomy and Embryology, University of Regensburg, Regensburg, Germany
| | - Preethi S. Ganapathy
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, Syracuse, New York, United States
| | - Haiyan Gong
- Department of Ophthalmology, Boston University School of Medicine, Boston, Massachusetts, United States
| | - Samuel Herberg
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, Syracuse, New York, United States
| | | | - Peter Humphries
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | - Simon W. M. John
- Department of Ophthalmology, Columbia University, New York, New York, United States
| | - Paul L. Kaufman
- Department of Ophthalmology and Visual Sciences, University of Wisconsin–Madison, Madison, Wisconsin, United States
| | - Kate E. Keller
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - Mary J. Kelley
- Department of Ophthalmology and Department of Integrative Biosciences, Oregon Health & Science University, Portland, Oregon, United States
| | - Ruth A. Kelly
- Ocular Genetics Unit, Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | - David Krizaj
- Department of Ophthalmology, University of Utah School of Medicine, Salt Lake City, Utah, United States
| | - Ajay Kumar
- Department of Ophthalmology, University of Pittsburgh, Pennsylvania, United States
| | - Brian C. Leonard
- Department of Surgical and Radiological Sciences, University of California, Davis, Davis, California, United States
| | - Raquel L. Lieberman
- Department of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, Georgia, United States
| | - Paloma Liton
- Department of Ophthalmology and Department of Pathology, Duke University, Durham, North Carolina, United States
| | - Yutao Liu
- Department of Cellular Biology and Anatomy, James & Jean Culver Vision Discovery Institute, Augusta University, Augusta, Georgia, United States
| | - Katy C. Liu
- Duke Eye Center, Duke Health, Durham, North Carolina, United States
| | - Navita N. Lopez
- Department of Neurobiology, University of Utah, Salt Lake City, Utah, United States
| | - Weiming Mao
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - Timur Mavlyutov
- Department of Ophthalmology and Visual Sciences, University of Wisconsin–Madison, Madison, Wisconsin, United States
| | - Fiona McDonnell
- Duke Eye Center, Duke Health, Durham, North Carolina, United States
| | - Gillian J. McLellan
- Department of Surgical Sciences and Department of Ophthalmology and Visual Sciences, University of Wisconsin–Madison, Madison, Wisconsin, United States
| | - Philip Mzyk
- Department of Ophthalmology and Visual Sciences, University of Wisconsin–Madison, Madison, Wisconsin, United States
| | - Andrews Nartey
- College of Optometry, University of Houston, Houston, Texas, United States
| | - Louis R. Pasquale
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, New York, United States
| | - Gaurang C. Patel
- Ophthalmology Research, Regeneron Pharmaceuticals, Tarreytown, New York, United States
| | | | - Donna M. Peters
- Department of Pathology and Laboratory Medicine, University of Wisconsin–Madison, Madison, Wisconsin, United States
| | | | - Ponugoti Vasantha Rao
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina, United States
| | - Naga Rayana
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - Urmimala Raychaudhuri
- Department of Neurobiology, University of California, Irvine, Irvine, California, United States
| | - Ester Reina-Torres
- Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Ruiyi Ren
- Department of Ophthalmology, Boston University School of Medicine, Boston, Massachusetts, United States
| | - Douglas Rhee
- Case Western Reserve University School of Medicine, Cleveland, Ohio, United States
| | - Uttio Roy Chowdhury
- Department of Ophthalmology, Mayo Clinic, Rochester, Minnesota, United States
| | - John R. Samples
- Washington State University, Floyd Elson College of Medicine, Spokane, Washington, United States
| | | | - Najam Sharif
- Santen Inc., Emeryville, California, United States
| | - Joel S. Schuman
- Department of Ophthalmology and Department of Physiology and Neuroscience, NYU Grossman School of Medicine, NYU Langone Health, New York University, New York, New York, United States; Departments of Biomedical Engineering and Electrical and Computer Engineering, New York University Tandon School of Engineering, Brooklyn, New York, United States; Center for Neural Science, College of Arts and Science, New York University, New York, New York, United States
| | - Val C. Sheffield
- Department of Pediatrics and Department of Ophthalmology and Visual Sciences, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States
| | - Cooper H. Stevenson
- Department of Pharmacology & Neuroscience, and North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Avinash Soundararajan
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | | | - Chenna Kesavulu Sugali
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - Yang Sun
- Veterans Affairs Palo Alto Health Care System, Stanford University, Palo Alto, California, United States
| | - Carol B. Toris
- Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, Nebraska, United States; Department of Ophthalmology and Vision Sciences, The Ohio State University, Columbus, Ohio, United States
| | | | - Amir Vahabikashi
- Cell and Developmental Biology Department, Northwestern University, Chicago, Illinois, United States
| | - Janice A. Vranka
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - Ting Wang
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - Colin E. Willoughby
- Genomic Medicine, Biomedical Sciences Research Institute, Ulster University, Coleraine, Northern Ireland, United Kingdom
| | - Chen Xin
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Hongmin Yun
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Hao F. Zhang
- Biomedical Engineering Department, Northwestern University, Evanston, Illinois, United States
| | - Michael P. Fautsch
- Biomedical Engineering Department, Northwestern University, Evanston, Illinois, United States
| | | | - Abbot F. Clark
- Department of Pharmacology and Neuroscience, North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - C. Ross Ethier
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology; Emory University School of Medicine, Emory University, Atlanta, Georgia, United States
| | - W. Daniel Stamer
- Duke Ophthalmology, Duke University, Durham, North Carolina, United States
| |
Collapse
|
14
|
Keller KE, Peters DM. Pathogenesis of glaucoma: Extracellular matrix dysfunction in the trabecular meshwork-A review. Clin Exp Ophthalmol 2022; 50:163-182. [PMID: 35037377 DOI: 10.1111/ceo.14027] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 11/19/2021] [Accepted: 11/26/2021] [Indexed: 12/11/2022]
Abstract
The trabecular meshwork regulates aqueous humour outflow from the anterior chamber of the eye. It does this by establishing a tunable outflow resistance, defined by the interplay between cells and their extracellular matrix (ECM) milieu, and the molecular interactions between ECM proteins. During normal tissue homeostasis, the ECM is remodelled and trabecular cell behaviour is modified, permitting increased aqueous fluid outflow to maintain intraocular pressure (IOP) within a relatively narrow physiological pressure. Dysfunction in the normal homeostatic process leads to increased outflow resistance and elevated IOP, which is a primary risk factor for glaucoma. This review delineates some of the changes in the ECM that lead to gross as well as some more subtle changes in the structure and function of the ECM, and their impact on trabecular cell behaviour. These changes are discussed in the context of outflow resistance and glaucoma.
Collapse
Affiliation(s)
- Kate E Keller
- Casey Eye Institute, Oregon Health &Science University, Portland, Oregon, USA
| | - Donna M Peters
- Department of Pathology & Laboratory Medicine, University of Wisconsin School of Medicine & Public Health, Madison, Wisconsin, USA
| |
Collapse
|
15
|
Relationships between Intraocular Pressure, Effective Filtration Area, and Morphological Changes in the Trabecular Meshwork of Steroid-Induced Ocular Hypertensive Mouse Eyes. Int J Mol Sci 2022; 23:ijms23020854. [PMID: 35055036 PMCID: PMC8775853 DOI: 10.3390/ijms23020854] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 01/06/2022] [Accepted: 01/10/2022] [Indexed: 02/04/2023] Open
Abstract
We investigated whether an inverse relationship exists between intraocular pressure (IOP) and effective filtration area (EFA) in the trabecular meshwork (TM) in a steroid-induced ocular hypertensive (SIOH) mouse model and the morphological changes associated with the reduction of EFA. C57BL/6 mice (n = 15 per group) received either 0.1% dexamethasone (DEX) or saline eye drops twice daily for five weeks. IOP was measured weekly. Fluorescent tracers were injected into the anterior chamber to label EFA at the endpoint. Injected eyes were fixed and processed for confocal microscopy. EFA in the TM was analyzed. Light and electron microscopy were performed in high- and low-tracer regions of six eyes per group. The mean IOP was ~4 mm Hg higher in DEX-treated than saline-treated control eyes (p < 0.001) at the endpoint. EFA was reduced in DEX-treated eyes compared to controls (p < 0.01) and negatively correlated with IOP (R2 = 0.38, p = 0.002). Reduced thickness of juxtacanalicular tissue (JCT) and increased abnormal extracellular matrix in the JCT were found to be associated with reduced EFA. Our data confirm the inverse relationship between EFA and IOP, suggesting that morphological changes in the JCT contribute to the reduction of EFA, thus elevating IOP in SIOH mouse eyes.
Collapse
|
16
|
Vincent MP, Stack T, Vahabikashi A, Li G, Perkumas KM, Ren R, Gong H, Stamer WD, Johnson M, Scott EA. Surface Engineering of FLT4-Targeted Nanocarriers Enhances Cell-Softening Glaucoma Therapy. ACS APPLIED MATERIALS & INTERFACES 2021; 13:32823-32836. [PMID: 34232612 PMCID: PMC9131393 DOI: 10.1021/acsami.1c09294] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Primary open-angle glaucoma is associated with elevated intraocular pressure (IOP) that damages the optic nerve and leads to gradual vision loss. Several agents that reduce the stiffness of pressure-regulating Schlemm's canal (SC) endothelial cells, in the conventional outflow pathway of the eye, lower IOP in glaucoma patients and are approved for clinical use. However, poor drug penetration and uncontrolled biodistribution limit their efficacy and produce local adverse effects. Compared to other ocular endothelia, FLT4/VEGFR3 is expressed at elevated levels by SC endothelial cells and can be exploited for targeted drug delivery. Here, we validate FLT4 receptors as clinically relevant targets on SC cells from glaucomatous human donors and engineer polymeric self-assembled nanocarriers displaying lipid-anchored targeting ligands that optimally engage this receptor. Targeting constructs were synthesized as lipid-PEGx-peptide, differing in the number of PEG spacer units (x), and were embedded in micelles. We present a novel proteolysis assay for quantifying ligand accessibility that we employ to design and optimize our FLT4-targeting strategy for glaucoma nanotherapy. Peptide accessibility to proteases correlated with receptor-mediated targeting enhancements. Increasing the accessibility of FLT4-binding peptides enhanced nanocarrier uptake by SC cells while simultaneously decreasing the uptake by off-target vascular endothelial cells. Using a paired longitudinal IOP study in vivo, we show that this enhanced targeting of SC cells translates to IOP reductions that are sustained for a significantly longer time as compared to controls. Confocal microscopy of murine anterior segment tissue confirmed nanocarrier localization to SC within 1 h after intracameral administration. This work demonstrates that steric effects between surface-displayed ligands and PEG coronas significantly impact the targeting performance of synthetic nanocarriers across multiple biological scales. Minimizing the obstruction of modular targeting ligands by PEG measurably improved the efficacy of glaucoma nanotherapy and is an important consideration for engineering PEGylated nanocarriers for targeted drug delivery.
Collapse
Affiliation(s)
- Michael P Vincent
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois 60208, United States
| | - Trevor Stack
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois 60208, United States
| | - Amir Vahabikashi
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois 60208, United States
- Department of Cell and Developmental Biology, Northwestern University, Chicago, Illinois 60611, United States
| | - Guorong Li
- Department of Ophthalmology, Duke University, Durham, North Carolina 27710, United States
| | - Kristin M Perkumas
- Department of Ophthalmology, Duke University, Durham, North Carolina 27710, United States
| | - Ruiyi Ren
- Department of Ophthalmology, Boston University School of Medicine, Boston, Massachusetts 02118, United States
| | - Haiyan Gong
- Department of Ophthalmology, Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, Massachusetts 02118, United States
| | - W Daniel Stamer
- Department of Ophthalmology, Duke University, Durham, North Carolina 27710, United States
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708, United States
| | - Mark Johnson
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois 60208, United States
- Department of Ophthalmology, Northwestern University, Chicago, Illinois 60611, United States
- Department of Mechanical Engineering, Northwestern University, Evanston, Illinois 60208, United States
| | - Evan A Scott
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois 60208, United States
- Simpson Querrey Institute, Northwestern University, Chicago, Illinois 60611, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois 60611, United States
| |
Collapse
|
17
|
Erlanger Glaucoma Registry: Effect of a Long-Term Therapy with Statins and Acetyl Salicylic Acid on Glaucoma Conversion and Progression. BIOLOGY 2021; 10:biology10060538. [PMID: 34208432 PMCID: PMC8234675 DOI: 10.3390/biology10060538] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 06/13/2021] [Accepted: 06/14/2021] [Indexed: 12/31/2022]
Abstract
Purpose: Drugs with cardiovascular protective properties (statins, acetylsalicylic acid (ASS)) were assumed to have positive effects on patients suffering from glaucoma disease. The present retrospective study aimed to investigate the influence of statins, ASS or a combination of both on the glaucoma conversion and progression rate in glaucoma suspects and glaucoma patients with a 20-year follow-up period. Methods: A retrospective analysis of 199 eyes of 120 patients (63 male, 57 female) of the Erlanger Glaucoma Registry (EGR; ClinicalTrials.gov Identifier: NCT00494923; ISSN 2191-5008, CS-2011) was performed considering systemic therapy with statins, ASS or a combination of both: 107 eyes with ocular hypertension (OHT) and 92 eyes with pre-perimetric primary open-angle glaucoma (pre-POAG). All patients received an ophthalmological examination including morphometric and functional glaucoma diagnostics. Glaucoma conversion was defined as the conversion of OHT to pre-POAG. Glaucoma progression was defined as confirmed visual field loss. Data were shown as percentages. Statistical analysis was performed by Chi-Quadrat tests. Results: 1. Glaucoma conversion/progression was observed in 46.7% of the subjects, additionally in combination with hypercholesterinemia in 76.8%. 2. Statins: 27.3% of eyes under systemic statin therapy showed a conversion/progression. Patients taking statins ≥ 10 years yielded a reduced conversion/progression rate (p = 0.028, non-significant after Bonferroni-Holm). 3. ASS: 34.7% of eyes under systemic ASS therapy showed a conversion/progression. A significantly lower conversion/progression rate was observed after ASS therapy ≥ 12 years (p = 0.017, significant after Bonferroni-Holm). 4. ASS and statins: 25.0% of eyes under combined therapy showed a conversion/progression. A significantly reduced conversion/progression rate was reached after 8 years of combined therapy (p = 0.049, non-significant after Bonferroni-Holm). Conclusions: Patients with ocular hypertension and early glaucoma seem to benefit from adjuvant cardiovascular protective therapy. However, the benefits and disadvantages of treatment with statins and/or ASS should be kept in mind. Thus, a thorough risk-benefit evaluation has to be performed for each patient individually to avoid unwanted side effects.
Collapse
|
18
|
Secreted protein acidic and rich in cysteine (SPARC) knockout mice have greater outflow facility. PLoS One 2020; 15:e0241294. [PMID: 33147244 PMCID: PMC7641442 DOI: 10.1371/journal.pone.0241294] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 10/12/2020] [Indexed: 11/24/2022] Open
Abstract
Purpose Secreted protein acidic and rich in cysteine (SPARC) is a matricellular protein that regulates intraocular pressure (IOP) by altering extracellular matrix (ECM) homeostasis within the trabecular meshwork (TM). We hypothesized that the lower IOP previously observed in SPARC -/- mice is due to a greater outflow facility. Methods Mouse outflow facility (Clive) was determined by multiple flow rate infusion, and episcleral venous pressure (Pe) was estimated by manometry. The animals were then euthanized, eliminating aqueous formation rate (Fin) and Pe. The C value was determined again (Cdead) while Fin was reduced to zero. Additional mice were euthanized for immunohistochemistry to analyze ECM components of the TM. Results The Clive and Cdead of SPARC -/- mice were 0.014 ± 0.002 μL/min/mmHg and 0.015 ± 0.002 μL/min/mmHg, respectively (p = 0.376, N/S). Compared to the Clive = 0.010 ± 0.002 μL/min/mmHg and Cdead = 0.011 ± 0.002 μL/min/mmHg in the WT mice (p = 0.548, N/S), the Clive and Cdead values for the SPARC -/- mice were higher. Pe values were estimated to be 8.0 ± 0.2 mmHg and 8.3 ± 0.7 mmHg in SPARC -/- and WT mice, respectively (p = 0.304, N/S). Uveoscleral outflow (Fu) was 0.019 ± 0.007 μL/min and 0.022 ± 0.006 μL/min for SPARC -/- and WT mice, respectively (p = 0.561, N/S). Fin was 0.114 ± 0.002 μL/min and 0.120 ± 0.016 μL/min for SPARC -/- and WT mice (p = 0.591, N/S). Immunohistochemistry demonstrated decreases of collagen types IV and VI, fibronectin, laminin, PAI-1, and tenascin-C within the TM of SPARC -/- mice (p < 0.05). Conclusions The lower IOP of SPARC -/- mice is due to greater aqueous humor outflow facility through the conventional pathway. Corresponding changes in several matricellular proteins and ECM structural components were noted in the TM of SPARC -/- mice.
Collapse
|
19
|
Lee JY, Akiyama G, Saraswathy S, Xie X, Pan X, Hong YK, Huang AS. Aqueous humour outflow imaging: seeing is believing. Eye (Lond) 2020; 35:202-215. [PMID: 33060830 DOI: 10.1038/s41433-020-01215-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 09/27/2020] [Accepted: 09/29/2020] [Indexed: 12/22/2022] Open
Abstract
Elevated intraocular pressure (IOP) is the primary risk factor for blindness in glaucoma. IOP is determined by many factors including aqueous humour production and aqueous humour outflow (AHO), where AHO disturbance represents the primary cause of increased IOP. With the recent development of new IOP lowering drugs and Minimally Invasive Glaucoma Surgeries (MIGS), renewed interest has arisen in shedding light on not only how but where AHO is occurring for the trabecular/conventional, uveoscleral/unconventional, and subconjunctival outflow pathways. Historical studies critical to understanding outflow anatomy will be presented, leading to the development of modern imaging methods. New biological behaviours uncovered by modern imaging methods will be discussed with relevance to glaucoma therapies emphasized.
Collapse
Affiliation(s)
- Jong Yeon Lee
- Doheny Eye Institute and Stein Eye Institute, Department of Ophthalmology, David Geffen School of Medicine, University of California, Los Angeles, California, USA.,Department of Ophthalmology, Gachon University, College of Medicine, Gil Medical Center, Incheon, Korea
| | - Goichi Akiyama
- Doheny Eye Institute and Stein Eye Institute, Department of Ophthalmology, David Geffen School of Medicine, University of California, Los Angeles, California, USA.,Jikei School of Medicine, Tokyo, Japan.,Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Sindhu Saraswathy
- Doheny Eye Institute and Stein Eye Institute, Department of Ophthalmology, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Xiaobin Xie
- Doheny Eye Institute and Stein Eye Institute, Department of Ophthalmology, David Geffen School of Medicine, University of California, Los Angeles, California, USA.,Eye Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaojing Pan
- Doheny Eye Institute and Stein Eye Institute, Department of Ophthalmology, David Geffen School of Medicine, University of California, Los Angeles, California, USA.,Qindao Eye Hospital of Shandong First Medical University, Shandong Eye Institute, Qindao, China
| | - Young-Kwon Hong
- Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Alex S Huang
- Doheny Eye Institute and Stein Eye Institute, Department of Ophthalmology, David Geffen School of Medicine, University of California, Los Angeles, California, USA.
| |
Collapse
|
20
|
Jeong JH, Lee SJ, Ko K, Lee JH, Lyu J, Park MH, Kang J, Kim JC. Plant-derived angiogenin fusion protein's cytoprotective effect on trabecular meshwork damage induced by Benzalkonium chloride in mice. PeerJ 2020; 8:e9084. [PMID: 32509448 PMCID: PMC7247531 DOI: 10.7717/peerj.9084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 04/08/2020] [Indexed: 11/20/2022] Open
Abstract
Background Benzalkonium chloride (BAK), commonly used in glaucoma treatment, is an eye drop preservative with dose-dependent toxicity. Previous studies have observed the multi-functional benefits of angiogenin (ANG) against glaucoma. In our study, we evaluated ANG’s cytoprotective effect on the trabecular meshwork (TM) damage induced by BAK. Additionally, we developed a plant-derived ANG fusion protein and evaluated its effect on TM structure and function. Methods We synthesized plant-derived ANG (ANG-FcK) by fuzing immunoglobulin G’s Fc region and KDEL to conventional recombinant human ANG (Rh-ANG) purified from transgenic tobacco plants. We established a mouse model using BAK to look for degenerative changes in the TM, and to evaluate the protective effects of ANG-FcK and Rh-ANG. Intraocular pressure (IOP) was measured for 4 weeks and ultrastructural changes, deposition of fluorescent microbeads, type I and IV collagen, fibronectin, laminin and α-SMA expression were analyzed after the mice were euthanized. Results TM structural and functional degeneration were induced by 0.1% BAK instillation in mice. ANG co-treatment preserved TM outflow function, which we measured using IOP and a microbead tracer. ANG prevented phenotypic and ultrastructure changes, and that protective effect might be related to the anti-fibrosis mechanism. We observed a similar cytoprotective effect in the BAK-induced degenerative TM mouse model, suggesting that plant-derived ANG-FcK could be a promising glaucoma treatment.
Collapse
Affiliation(s)
- Jae Hoon Jeong
- Department of Ophthalmology, Konyang University Hospital, Daejeon, South Korea.,Myunggok Medical Research Institute, Konyang University, Daejeon, South Korea.,Myunggok Eye Research Institute, Konyang University, Daejeon, South Korea
| | - Soo Jin Lee
- Department of Ophthalmology, Chung-Ang University Hospital, Seoul, South Korea
| | - Kisung Ko
- Therapeutic Protein Engineering Lab/College of Medicine, Chung-Ang University, Seoul, South Korea
| | - Jeong Hwan Lee
- Therapeutic Protein Engineering Lab/College of Medicine, Chung-Ang University, Seoul, South Korea
| | - Jungmook Lyu
- Myunggok Eye Research Institute, Konyang University, Daejeon, South Korea.,Department of Medical Science, Konyang University, Daejeon, South Korea
| | - Moon Hyang Park
- Department of Pathology, Konyang University Hospital, Daejeon, South Korea
| | - Jaeku Kang
- Myunggok Medical Research Institute, Konyang University, Daejeon, South Korea.,Department of Pharmacology/College of Medicine, Konyang University, Daejeon, South Korea
| | - Jae Chan Kim
- Department of Ophthalmology, Chung-Ang University Hospital, Seoul, South Korea
| |
Collapse
|
21
|
Secreted Protein Acidic and Rich in Cysteine: Metabolic and Homeostatic Properties beyond the Extracellular Matrix Structure. APPLIED SCIENCES-BASEL 2020. [DOI: 10.3390/app10072388] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
An extracellular matrix (ECM) is a network of numerous macromolecules that represents the cellular structural support involved in key biofunctions such as signal transduction and cellular adhesion. In addition, ECM-associated proteins interact with ECM and with other endogenous structures and molecules to control cellular growth, structural modifications, cellular migration, etc. Among the ECM-associated proteins, secreted protein acidic and rich in cysteine (SPARC) is a protein that is known to be expressed when tissues change. Herein, we put a spotlight on selected, metabolic and homeostatic properties beyond the known properties of ECM and SPARC. Importantly, the synchronization of the metabolic and structural implications of SPARC and the ECM would indicate an adaptation of the metabolism to meet the needs of the changes that the tissues undergo. Highlighting such properties would have important applications in diverse fields that include therapeutics, metabolics, and pathogenesis.
Collapse
|
22
|
NikhalaShree S, Karthikkeyan G, George R, Shantha B, Vijaya L, Ratra V, Sulochana KN, Coral K. Lowered Decorin With Aberrant Extracellular Matrix Remodeling in Aqueous Humor and Tenon's Tissue From Primary Glaucoma Patients. Invest Ophthalmol Vis Sci 2020; 60:4661-4669. [PMID: 31725165 DOI: 10.1167/iovs.19-27091] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose To evaluate the inflammatory cytokine, growth factors, extracellular matrix (ECM) remodeling genes, profibrotic and antifibrotic molecules in patients undergoing glaucoma filtration surgery (GFS). Additionally, the effect of preoperative antiglaucoma medications (AGMs) and postoperative bleb status were related to these parameters. Methods Tenon's tissue and aqueous humour (AH) were collected from 207 patients undergoing GFS with primary open-angle glaucoma (POAG) (n = 77), primary angle-closure glaucoma (PACG) (n = 62), and cataract controls (n = 68). Monocyte chemoattractant protein-1 (MCP-1), connective tissue growth factor (CTGF), transforming growth factor β1/2 (TGF-β1/2), lysyl oxidase (LOX), lysyl oxidase L2 (LOXL2), elastin (ELN), collagen type 1 α 1 (COL1A1), secreted protein acidic and rich in cysteine (SPARC), α-smooth muscle actin (α-SMA), and decorin (DCN) were determined in tenon's tissue by real-time PCR and in AH using ELISA. Results A significant increase was observed in the transcripts of MCP-1, TGF-β2, and SPARC in POAG and PACG (P < 0.05); CTGF, TGF-β1, LOX, LOXL2, ELN, COL1A1, and α-SMA in PACG (P < 0.05) compared with control. DCN transcript was significantly decreased in POAG and PACG (P < 0.05) compared with control. The protein levels of CTGF, TGF-β1/β2, ELN, SPARC, and LOXL2 was significantly elevated in POAG and PACG (P < 0.05); DCN was decreased (P < 0.05) compared with control. These parameters showed significant association with duration of preoperative AGMs and postoperative bleb status. Conclusions This study demonstrates increased expression of growth factors and ECM molecules, both at protein and transcript levels in GFS patients. A decreased DCN in AH seems striking, and if restored might have a therapeutic role in minimizing postoperative scarring to improve GFS outcome.
Collapse
Affiliation(s)
- Sampath NikhalaShree
- R.S. Mehta Jain Department of Biochemistry and Cell Biology, KBIRVO, Vision Research Foundation, Sankara Nethralaya, Chennai, India.,School of Chemical and Biotechnology, SASTRA Deemed-to-be University, Thanjavur, India
| | - Gayathree Karthikkeyan
- R.S. Mehta Jain Department of Biochemistry and Cell Biology, KBIRVO, Vision Research Foundation, Sankara Nethralaya, Chennai, India
| | - Ronnie George
- Smt Jadhavbai Nathmal Singhvee Glaucoma Services, Medical Research Foundation, Sankara Nethralaya, Chennai, India
| | - Balekudaru Shantha
- Smt Jadhavbai Nathmal Singhvee Glaucoma Services, Medical Research Foundation, Sankara Nethralaya, Chennai, India
| | - Lingam Vijaya
- Smt Jadhavbai Nathmal Singhvee Glaucoma Services, Medical Research Foundation, Sankara Nethralaya, Chennai, India
| | - Vineet Ratra
- Smt Jadhavbai Nathmal Singhvee Glaucoma Services, Medical Research Foundation, Sankara Nethralaya, Chennai, India
| | | | - Karunakaran Coral
- R.S. Mehta Jain Department of Biochemistry and Cell Biology, KBIRVO, Vision Research Foundation, Sankara Nethralaya, Chennai, India
| |
Collapse
|
23
|
Episcleral Venous Fluid Wave in the Living Human Eye Adjacent to Microinvasive Glaucoma Surgery (MIGS) Supports Laboratory Research: Outflow is Limited Circumferentially, Conserved Distally, and Favored Inferonasally. J Glaucoma 2020; 28:139-145. [PMID: 30461548 DOI: 10.1097/ijg.0000000000001126] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE The purpose of this study was to describe downstream patterns of outflow with the episcleral venous fluid wave (EVFW) in the living human eye adjacent to microinvasive glaucoma surgery (MIGS) and determine if the EVFW supports existing ex-vivo laboratory outflow research. DESIGN Retrospective, noncomparative case series. PATIENTS A total of 10 eyes of 10 patients who underwent phaco-Trabectome and 10 eyes of 10 patients who underwent phaco-iStent consecutively at Glaucoma Associates of Texas for cataract and uncontrolled glaucoma who demonstrated an episcleral wave. METHODS The EVFW was visualized and recorded during irrigation and aspiration. To describe the hydrodynamic properties of the fluid wave, its degrees, extent, and characteristics were measured with a protractor in Photoshop. RESULTS The incised Trabectome arc produced adjacent episcleral blanching of 134±11 degrees (range, 112 to 150 degrees) with an additional 54 degrees of marginal recruitment (41 degrees inferonasal plus 13 degrees superonasal) adjacent to the ends of the Trabectome incision. The mean episcleral blanch for the iStent was 51±19 degrees (range, 19 to 90 degrees), comprised of 29 degrees inferonasal plus 22 degrees superonasal. CONCLUSIONS Downstream episcleral flow in the living human eye adjacent to the iStent is variable and mainly confined to 2 clock hours indicating a lack of significant circumferential flow in glaucomatous eyes. Flow distal to the Trabectome site encompasses the Trabectome incisional arc with an additional 2 clock hours of lateral fluid wave favoring the inferonasal over superonasal quadrant 3 to 1. These in-vivo findings made visible with MIGS, corroborate recent in-vivo and long-standing ex-vivo laboratory research that outflow is largely segmented, favored inferonasally and conserved distally.
Collapse
|
24
|
How many aqueous humor outflow pathways are there? Surv Ophthalmol 2019; 65:144-170. [PMID: 31622628 DOI: 10.1016/j.survophthal.2019.10.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Revised: 09/29/2019] [Accepted: 10/04/2019] [Indexed: 12/31/2022]
Abstract
The aqueous humor (AH) outflow pathways definition is still matter of intense debate. To date, the differentiation between conventional (trabecular meshwork) and unconventional (uveoscleral) pathways is widely accepted, distinguishing the different impact of the intraocular pressure on the AH outflow rate. Although the conventional route is recognized to host the main sites for intraocular pressure regulation, the unconventional pathway, with its great potential for AH resorption, seems to act as a sort of relief valve, especially when the trabecular resistance rises. Recent evidence demonstrates the presence of lymphatic channels in the eye and proposes that they may participate in the overall AH drainage and intraocular pressure regulation, in a presumably adaptive fashion. For this reason, the uveolymphatic route is increasingly thought to play an important role in the ocular hydrodynamic system physiology. As a result of the unconventional pathway characteristics, hydrodynamic disorders do not develop until the adaptive routes cannot successfully counterbalance the increased AH outflow resistance. When their adaptive mechanisms fail, glaucoma occurs. Our review deals with the standard and newly discovered AH outflow routes, with particular attention to the importance they may have in opening new therapeutic strategies in the treatment of ocular hypertension and glaucoma.
Collapse
|
25
|
Snyder KC, Oikawa K, Williams J, Kiland JA, Gehrke S, Teixeira LBC, Huang AS, McLellan GJ. Imaging Distal Aqueous Outflow Pathways in a Spontaneous Model of Congenital Glaucoma. Transl Vis Sci Technol 2019; 8:22. [PMID: 31616579 PMCID: PMC6788461 DOI: 10.1167/tvst.8.5.22] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 07/26/2019] [Indexed: 01/01/2023] Open
Abstract
PURPOSE To validate the use of aqueous angiography (AA) in characterizing distal aqueous outflow pathways in normal and glaucomatous cats. METHODS Ex vivo AA and optical coherence tomography (OCT) were performed in nine adult cat eyes (5 feline congenital glaucoma [FCG] and 4 normal), following intracameral infusion of 2.5% fluorescein and/or 0.4% indocyanine green (ICG) at physiologic intraocular pressure (IOP). Scleral OCT line scans were acquired in areas of high- and low-angiographic signal. Tissues dissected in regions of high- and low-AA signal, were sectioned and hematoxylin and eosin (H&E)-stained or immunolabeled (IF) for vascular endothelial and perivascular cell markers. Outflow vessel numbers and locations were compared between groups by Student's t-test. RESULTS AA yielded circumferential, high-quality images of distal aqueous outflow pathways in normal and FCG eyes. No AA signal or scleral lumens were appreciated in one buphthalmic FCG eye, though collapsed vascular profiles were identified on IF. The remaining eight of nine eyes all showed segmental AA signal, distinguished by differences in time of signal onset. AA signal always corresponded with lumens seen on OCT. Numbers of intrascleral vessels were not significantly different between groups, but scleral vessels were significantly more posteriorly located relative to the limbus in FCG. CONCLUSIONS A capacity for distal aqueous humor outflow was confirmed by AA in FCG eyes ex vivo but with significant posterior displacement of intrascleral vessels relative to the limbus in FCG compared with normal eyes. TRANSLATIONAL RELEVANCE This report provides histopathologic correlates of advanced diagnostic imaging findings in a spontaneous model of congenital glaucoma.
Collapse
Affiliation(s)
- Kevin C. Snyder
- Department of Surgical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, WI, USA
| | - Kazuya Oikawa
- Department of Surgical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, WI, USA
- Department of Ophthalmology and Visual Sciences, School of Medicine and Public Health, University of Wisconsin-Madison, WI, USA
| | - Jeremy Williams
- Department of Ophthalmology and Visual Sciences, School of Medicine and Public Health, University of Wisconsin-Madison, WI, USA
| | - Julie A. Kiland
- Department of Ophthalmology and Visual Sciences, School of Medicine and Public Health, University of Wisconsin-Madison, WI, USA
| | - Shaile Gehrke
- Department of Surgical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, WI, USA
| | - Leandro B. C. Teixeira
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, WI, USA
| | - Alex S. Huang
- Doheny Eye Institute, and Department of Ophthalmology University of California, Los Angeles, CA, USA
| | - Gillian J. McLellan
- Department of Surgical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, WI, USA
- Department of Ophthalmology and Visual Sciences, School of Medicine and Public Health, University of Wisconsin-Madison, WI, USA
| |
Collapse
|
26
|
Xie X, Akiyama G, Bogarin T, Saraswathy S, Huang AS. Visual Assessment of Aqueous Humor Outflow. Asia Pac J Ophthalmol (Phila) 2019; 8:126-134. [PMID: 30916496 PMCID: PMC7028348 DOI: 10.22608/apo.201911] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
In the past decade, many new pharmacological and surgical treatments have become available to lower intraocular pressure (IOP) for glaucoma. The majority of these options have targeted improving aqueous humor outflow (AHO). At the same time, in addition to new treatments, research advances in AHO assessment have led to the development of new tools to structurally assess AHO pathways and to visualize where aqueous is flowing in the eye. These new imaging modalities have uncovered novel AHO observations that challenge traditional AHO concepts. New behaviors including segmental, pulsatile, and dynamic AHO may have relevance to the disease and the level of therapeutic response for IOP-lowering treatments. By better understanding the regulation of segmental, pulsatile, and dynamic AHO, it may be possible to find new and innovative treatments for glaucoma aiming at these new AHO behaviors.
Collapse
Affiliation(s)
- Xiaobin Xie
- From the Eye Hospital of China Academy of Chinese Medical Sciences, Beijing, China; and UCLA Department of Ophthalmology, Doheny Eye Institute, Los Angeles, CA, United States
| | | | | | | | | |
Collapse
|
27
|
Jiang Y, Zhang C, Ma J, Wang L, Gao J, Ren J, He W, Wang S, Sheng S, Huang X. Expression of matrix Metalloproteinases-2 and aquaporin-1 in corneoscleral junction after angle-closure in rabbits. BMC Ophthalmol 2019; 19:43. [PMID: 30717683 PMCID: PMC6360660 DOI: 10.1186/s12886-019-1058-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 01/29/2019] [Indexed: 11/24/2022] Open
Abstract
Background To investigate the expression of Matrix Metalloproteinases 2 and aquaporin-1 in corneoscleral junction and explore the mechanism of trabecular damageafter angle-closure. Methods Thirty New Zealand white rabbits were randomly assigned into 2 groups, theexperimental group (Group 1) including twenty five rabbits and the control group (Group 2) including 5 rabbits. The rabbits in the experimental group were used to establish angle-closure models, and the rabbits in the control group were not subjected to any operation. All the rabbits were followed by slit lamp microscopy, Tonopen tonometer, and anterior segment optical coherent tomography (AS-OCT). The expressions of metalloproteinase MMP-2, aquaporin-1, and tissue inhibitors of metalloproteinase-2 in corneoscleral junctionwere evaluatedin both groups byimmunofluorescence, quantitative reverse-transcription polymerase chain reaction (qRT-PCR), and enzyme-linked immunosorbent assay (ELISA). Results Slit-lamp examination showed that angle-closure model was successfully established in twenty rabbits. The extent of angle-closure was about 2 to 4 clock hours in all the rabbit models, but the intraocular pressure (IOP) of the rabbits distributed from 8.57 to 15.25 mmHg and no significant high IOP was found in the follow-up period. The AQP-1-positive cells mainly located in Schlemm’s canal, the inner surface of trabecular meshwork (TM), and the surface of iris, which began to decline on 1 month after angle-closure. MMP2 staining was diffuse in trabecular meshwork and iris. Immunofluorescence signal of MMP2 was strong within 1 month after angle-closure, and subsequently became weak. qRT-PCR and ELISA showed that the expression of MMP-2 and TIMP-2 increased within 1 month after angle-closure and then declined gradually. The AQP-1 levels showed slightly declined on 1 month after angle-closure. Conclusions Altered levels of MMPs, TIMPs, and AQP-1 were found in the area of angle-closure, which may be involved in the damage of TM and Schlemm’s canal after angle-closure.
Collapse
Affiliation(s)
- Yaqin Jiang
- Department of Ophthalmology, Weifang Eye Hospital, Weifang, Shandong, People's Republic of China
| | - Canwei Zhang
- Department of Ophthalmology, Weifang Eye Hospital, Weifang, Shandong, People's Republic of China
| | - Jianli Ma
- Department of Ophthalmology, Weifang Eye Hospital, Weifang, Shandong, People's Republic of China
| | - Luping Wang
- Department of Ophthalmology, University of Bonn, Bonn, Germany
| | - Jing Gao
- Department of Ophthalmology, Weifang Eye Hospital, Weifang, Shandong, People's Republic of China
| | - Jiantao Ren
- Department of Ophthalmology, Weifang Eye Hospital, Weifang, Shandong, People's Republic of China
| | - Wei He
- Department of Ophthalmology, Weifang Eye Hospital, Weifang, Shandong, People's Republic of China
| | - Sheng Wang
- Department of Ophthalmology, Weifang Eye Hospital, Weifang, Shandong, People's Republic of China
| | - Shuai Sheng
- Department of Ophthalmology, Weifang Eye Hospital, Weifang, Shandong, People's Republic of China
| | - Xudong Huang
- Department of Ophthalmology, Weifang Eye Hospital, Weifang, Shandong, People's Republic of China.
| |
Collapse
|
28
|
Huang AS, Penteado RC, Papoyan V, Voskanyan L, Weinreb RN. Aqueous Angiographic Outflow Improvement after Trabecular Microbypass in Glaucoma Patients. Ophthalmol Glaucoma 2018; 2:11-21. [PMID: 31595267 DOI: 10.1016/j.ogla.2018.11.010] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Purpose To study changes in aqueous humor outflow (AHO) patterns after trabecular micro-bypass (TMB) in glaucoma patients using intraoperative sequential aqueous angiography. Design Prospective comparative case series. Subjects Fifteen subjects (14 with glaucoma and 1 normal). Methods Sequential aqueous angiography (Spectralis HRA+OCT; Heidelberg Engineering) was performed on fourteen glaucoma patients undergoing routine TMB (iStent Inject; Glaukos Corporation) and cataract surgery and one normal patient undergoing cataract surgery alone. Indocyanine green (ICG) aqueous angiography established initial baseline nasal angiographic AHO patterns. Two TMB stents were placed in regions of baseline low or high angiographic AHO in each eye (n = 2 eyes with enough space to place two stents in both low angiographic regions; n = 8 eyes with two stents both placed in high angiographic regions; n = 4 eyes with enough space to place one stent in a low angiographic region and the other stent in a high angiographic region). Subsequent fluorescein aqueous angiography was utilized to query alterations to angiographic AHO patterns. Main Outcome Measure Angiographic signal and patterns before and after TMB. Results At baseline, all eyes showed segmental angiographic AHO patterns. Focused on the nasal hemisphere of each eye, for each stent TMB in initially low ICG angiographic signal regions showed transient or persistently improved fluorescein angiographic signal (11.2-fold; p = 0.014). TMB in initially high ICG signal regions led to faster development of fluorescein angiographic patterns (3.1-fold; p = 0.02). Conclusion TMB resulted in different patterns of aqueous angiographic AHO improvement whose further understanding may advance basic knowledge of AHO and possibly enhance intraocular pressure reduction after glaucoma surgery in the future.
Collapse
Affiliation(s)
- Alex S Huang
- Doheny Eye Institute and Department of Ophthalmology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Rafaella C Penteado
- Hamilton Glaucoma Center, Shiley Eye Institute, and Department of Ophthalmology University of California, San Diego, CA, USA
| | - Vahan Papoyan
- Department of Ophthalmology, Yerevan State Medical University, Ophthalmological Center after S.V. Malayan
| | - Lilit Voskanyan
- Department of Ophthalmology, Yerevan State Medical University, Ophthalmological Center after S.V. Malayan
| | - Robert N Weinreb
- Hamilton Glaucoma Center, Shiley Eye Institute, and Department of Ophthalmology University of California, San Diego, CA, USA
| |
Collapse
|
29
|
Huang AS, Penteado RC, Saha SK, Do JL, Ngai P, Hu Z, Weinreb RN. Fluorescein Aqueous Angiography in Live Normal Human Eyes. J Glaucoma 2018; 27:957-964. [PMID: 30095604 PMCID: PMC6218293 DOI: 10.1097/ijg.0000000000001042] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE To evaluate aqueous humor outflow (AHO) in intact eyes of live human subjects during cataract surgery using fluorescein aqueous angiography. METHODS Aqueous angiography was performed in 8 live human subjects (56 to 86 y old; 2 men and 6 women). After anesthesia, fluorescein (2%) was introduced into the eye [either alone or after indocyanine green (ICG; 0.4%)] from a sterile, gravity-driven constant-pressure reservoir. Aqueous angiographic images were obtained with a Spectralis HRA+OCT and FLEX module (Heidelberg Engineering). Using the same device, anterior-segment optical coherence tomography (OCT) and infrared images were also concurrently taken with aqueous angiography. RESULTS Fluorescein aqueous angiography in the live human eye showed segmental AHO patterns. Initial angiographic signal was seen on average by 14.0±3.0 seconds (mean±SE). Using multimodal imaging, angiographically positive signal colocalized with episcleral veins (infrared imaging) and intrascleral lumens (anterior-segment OCT). Sequential aqueous angiography with ICG followed by fluorescein showed similar segmental angiographic patterns. DISCUSSION Fluorescein aqueous angiography in live humans was similar to that reported in nonhuman primates and to ICG aqueous angiography in live humans. As segmental patterns with sequential angiography using ICG followed by fluorescein were similar, these tracers can now be used sequentially, before and after trabecular outflow interventions, to assess their effects on AHO in live human subjects.
Collapse
Affiliation(s)
- Alex S Huang
- Department of Ophthalmology, Doheny Eye Institute, David Geffen School of Medicine at UCLA, Los Angeles
| | - Rafaella C Penteado
- Department of Ophthalmology, Hamilton Glaucoma Center, Shiley Eye Institute, University of California, San Diego, CA
| | - Sajib K Saha
- Department of Ophthalmology, Doheny Eye Institute, David Geffen School of Medicine at UCLA, Los Angeles
| | - Jiun L Do
- Department of Ophthalmology, Hamilton Glaucoma Center, Shiley Eye Institute, University of California, San Diego, CA
| | - Philip Ngai
- Department of Ophthalmology, Hamilton Glaucoma Center, Shiley Eye Institute, University of California, San Diego, CA
| | - Zhihong Hu
- Department of Ophthalmology, Doheny Eye Institute, David Geffen School of Medicine at UCLA, Los Angeles
| | - Robert N Weinreb
- Department of Ophthalmology, Hamilton Glaucoma Center, Shiley Eye Institute, University of California, San Diego, CA
| |
Collapse
|
30
|
Swaminathan SS, Monsalve P, Zhou XY, Enriquez-Algeciras M, Bhattacharya SK, Dubovy SR, Junk AK. Histologic Analysis of Trabecular Meshwork Obtained From Kahook Dual Blade Goniotomy. Am J Ophthalmol 2018; 192:198-205. [PMID: 29883587 DOI: 10.1016/j.ajo.2018.05.028] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 05/24/2018] [Accepted: 05/29/2018] [Indexed: 12/13/2022]
Abstract
PURPOSE To determine whether there are identifiable, reproducible findings in the trabecular meshwork (TM) of patients with primary open-angle glaucoma (POAG) who underwent Kahook Dual Blade (KDB) goniotomy. DESIGN Noncomparative retrospective case series. METHODS Tertiary academic referral center, Veterans Affairs Medical Center. Thirteen patients (14 eyes) with POAG (100%) were treated with KDB goniotomy from May to December 2017. Isolated TM tissue was collected from 9 patients (10 eyes) and submitted for histologic analysis. Hematoxylin-eosin, periodic acid-Schiff, and elastin Van Gieson stains were completed, in addition to immunohistochemistry for collagen IV. RESULTS Mean age of patients was 74.2 ± 6.7 years. Trabecular beams were identified in all 10 specimens, although distorted in 4 samples, of which 3 had a history of laser trabeculoplasty. Collagen IV staining was present in 10 of 10 samples, coating the trabecular beams. Elastin was present in 8 of 10 samples along the trabecular beams. Intraocular pressure and number of glaucoma medications decreased significantly in all cases postoperatively (P < .0001, P = .035, respectively). CONCLUSIONS This pilot study demonstrates that tissue obtained during KDB goniotomy has a high yield of containing TM compared to reported yield of TM in specimens collected from traditional ab externo trabeculectomy (71% vs 20%, respectively). These goniotomy specimens possess sufficient anatomic preservation to be studied histologically. Trabecular meshwork obtained with this procedure may provide a novel modality to study TM dysfunction in open-angle glaucomas.
Collapse
Affiliation(s)
- Swarup S Swaminathan
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Pedro Monsalve
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Xiao Yi Zhou
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Mabel Enriquez-Algeciras
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Sanjoy K Bhattacharya
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Sander R Dubovy
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA; Florida Lions Ocular Pathology Laboratory, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Anna K Junk
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA; Miami Veterans Affairs Healthcare System, Miami, Florida, USA.
| |
Collapse
|
31
|
Vranka JA, Staverosky JA, Reddy AP, Wilmarth PA, David LL, Acott TS, Russell P, Raghunathan VK. Biomechanical Rigidity and Quantitative Proteomics Analysis of Segmental Regions of the Trabecular Meshwork at Physiologic and Elevated Pressures. Invest Ophthalmol Vis Sci 2018; 59:246-259. [PMID: 29340639 PMCID: PMC5770183 DOI: 10.1167/iovs.17-22759] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Purpose The extracellular matrix (ECM) of the trabecular meshwork (TM) modulates resistance to aqueous humor outflow, thereby regulating IOP. Glaucoma, a leading cause of irreversible blindness worldwide, is associated with changes in the ECM of the TM. The elastic modulus of glaucomatous TM is larger than age-matched normal TM; however, the biomechanical properties of segmental low (LF) and high flow (HF) TM regions and their response to elevated pressure, are unknown. Methods We perfused human anterior segments at two pressures using an ex vivo organ culture system. After extraction, we measured the elastic modulus of HF and LF TM regions by atomic force microscopy and quantitated protein differences by proteomics analyses. Results The elastic modulus of LF regions was 2.3-fold larger than HF regions at physiological (1×) pressure, and 7.4-fold or 3.5-fold larger than HF regions at elevated (2×) pressure after 24 or 72 hours, respectively. Using quantitative proteomics, comparisons were made between HF and LF regions at 1× or 2× pressure. Significant ECM protein differences were observed between LF and HF regions perfused at 2×, and between HF regions at 1× compared to 2× pressures. Decorin, TGF-β–induced protein, keratocan, lumican, dermatopontin, and thrombospondin 4 were common differential candidates in both comparisons. Conclusions These data show changes in biomechanical properties of segmental regions within the TM in response to elevated pressure, and levels of specific ECM proteins. Further studies are needed to determine whether these ECM proteins are specifically involved in outflow resistance and IOP homeostasis.
Collapse
Affiliation(s)
- Janice A Vranka
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - Julia A Staverosky
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - Ashok P Reddy
- Proteomics Shared Resources, Oregon Health & Science University, Portland, Oregon, United States
| | - Phillip A Wilmarth
- Proteomics Shared Resources, Oregon Health & Science University, Portland, Oregon, United States.,Department of Biochemistry and Molecular Biology, Oregon Health & Science University, Portland, Oregon, United States
| | - Larry L David
- Proteomics Shared Resources, Oregon Health & Science University, Portland, Oregon, United States.,Department of Biochemistry and Molecular Biology, Oregon Health & Science University, Portland, Oregon, United States
| | - Ted S Acott
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States.,Department of Biochemistry and Molecular Biology, Oregon Health & Science University, Portland, Oregon, United States
| | - Paul Russell
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California-Davis, Davis, California, United States
| | - Vijay Krishna Raghunathan
- Department of Basic Sciences, The Ocular Surface Institute, College of Optometry, University of Houston, Houston, Texas, United States.,Department of Biomedical Engineering, Cullen College of Engineering, University of Houston, Houston, Texas, United States
| |
Collapse
|
32
|
Wang C, Li L, Liu Z. Experimental research on the relationship between the stiffness and the expressions of fibronectin proteins and adaptor proteins of rat trabecular meshwork cells. BMC Ophthalmol 2017; 17:268. [PMID: 29284449 PMCID: PMC5747132 DOI: 10.1186/s12886-017-0662-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 12/12/2017] [Indexed: 12/28/2022] Open
Abstract
Background Trabecular meshwork (TM) plays an important role in maintaining normal intraocular pressure (IOP). Studies have shown that glaucomatous TM tissues are stiffer than those of normal tissue. The high expression of fibronectin protein (FN) and adaptor protein (LNK) may be related to high resistance to aqueous humor outflow as well as high IOP. Our concern is what factors lead to the variation of the stiffness of trabecular tissue/cells. Methods Atomic force microscope (AFM) and Western blot (WB) analysis were applied to test TM cells of rats cultured with different concentrations of dexamethasone (DEX) and mifepristone (MIF). Rat TM cells were randomly divided into 7 groups, marked as D1, D2, D3 and M1, M2 M3 for different concentrations of DEX and MIF, respectively, and C for blank control. Results The elastic modulus of the treated cells were 2.67 ± 0.914 KPa, 2.92 ± 0.986 KPa, 4.52 ± 1.22 KPa for D1, D2, D3, 2.06 ± 0.745 KPa, 1.23 ± 0.462 KPa, 0.467 ± 0.275 KPa for M1, M2, M3, and 2.43 ± 0.713 KPa for C group, respectively. Expressions of FN and LNK increase (decrease) with the increase of the concentrations of DEX (MIF). Discussion We focus on the relationship between the stiffness and the expressions of FN and LNK of rat TM cells. We analyzed the correlation between cell stiffness and FN, LNK expression, discussed the relationship between cell stiffness and aqueous humor outflow resistance. Conclusions The changes of TM cell stiffness and the expressions of FN and LNK are positively correlated.
Collapse
Affiliation(s)
- Chuan Wang
- Department of Biomechanics and Rehabilitation Engineering, School of Biomedical Engineering, Capital Medical University, No.10 Xitoutiao, You An Men, Beijing, 100069, People's Republic of China.,Beijing Key Laboratory of Fundamental Research on Biomechanics in Clinical Application, Capital Medical University, Beijing, 100069, China.,YanJing Medical College, Capital Medical University, Beijing, 100069, China
| | - Lin Li
- Department of Biomedical Informatics, School of Biomedical Engineering, Capital Medical University, No.10 Xitoutiao, You An Men, Beijing, 100069, People's Republic of China.
| | - Zhicheng Liu
- Department of Biomechanics and Rehabilitation Engineering, School of Biomedical Engineering, Capital Medical University, No.10 Xitoutiao, You An Men, Beijing, 100069, People's Republic of China. .,Beijing Key Laboratory of Fundamental Research on Biomechanics in Clinical Application, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
33
|
Wecker T, van Oterendorp C, Reichardt W. Functional assessment of the aqueous humour distal outflow pathways in bovine eyes using time-of-flight magnetic resonance tomography. Exp Eye Res 2017; 166:168-173. [PMID: 29074388 DOI: 10.1016/j.exer.2017.10.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 10/16/2017] [Accepted: 10/16/2017] [Indexed: 11/29/2022]
Abstract
The major part of the aqueous humor leaves the eye through the "conventional outflow pathway", consisting of the trabecular meshwork, Schlemm's canal, collector channels, an intrasceral plexus and the episcleral veins. While the trabecular meshwork is well characterized, little is known about anatomical and functional features of the peripheral outflow tract beyond Schlemm's canal. The emergence of minimally-invasive glaucoma surgery directly targeting the outflow resistance in the trabecular meshwork has elicited growing interest in these structures. We used time-of-flight magnetic resonance imaging in ex vivo bovine eyes to map fluid flow under physiological conditions. We were able to identify the peripheral outflow vessels solely by the signal detected from the fluid flow inside their lumina. A question of clinical relevance is, whether localized opening of the trabecular meshwork leads to only localized or to a 360° increase in intrascleral flow. To address this, a goniotomy ab interno was performed in 3 eyes and the flow signal intensity was quantified sectorially. A significant increase in fluid flow was observed in the sector distal to the goniotomy (p = 0.0005) but not in the other sectors (p = 0.1). As a proof of concept we demonstrated that TOF-MRI based detection of flow in the peripheral aqueous outflow tract is feasible. The functional link observed between trabecular meshwork sectors and their distal outflow tract sectors may be relevant for minimally-invasive glaucoma surgery in humans.
Collapse
Affiliation(s)
- Thomas Wecker
- Eye Center, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany; Wecker Eye Center, Heilbronn, Germany.
| | | | - Wilfried Reichardt
- Medical Physics, Department of Radiology, Faculty of Medicine, University of Freiburg, Germany; German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
34
|
Huang AS, Francis BA, Weinreb RN. Structural and functional imaging of aqueous humour outflow: a review. Clin Exp Ophthalmol 2017; 46:158-168. [PMID: 28898516 DOI: 10.1111/ceo.13064] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 09/06/2017] [Indexed: 01/15/2023]
Abstract
Maintaining healthy aqueous humour outflow (AHO) is important for intraocular cellular health and stable vision. Impairment of AHO can lead to increased intraocular pressure, optic nerve damage and concomitant glaucoma. An improved understanding of AHO will lead to improved glaucoma surgeries that enhance native AHO as well as facilitate the development of AHO-targeted pharmaceuticals. Recent AHO imaging has evolved to live human assessment and has focused on the structural evaluation of AHO pathways and the functional documentation of fluid flow. Structural AHO evaluation is predominantly driven by optical coherence tomography, and functional evaluation of flow is performed using various methods, including aqueous angiography. Advances in structural and functional evaluation of AHO are reviewed with discussion of strengths, weaknesses and potential future directions.
Collapse
Affiliation(s)
- Alex S Huang
- Doheny Eye Institute, Los Angeles, California, USA.,Doheny Eye Centers, Department of Ophthalmology, David Geffen School of Medicine at University of California, Los Angeles, California, USA
| | - Brian A Francis
- Doheny Eye Institute, Los Angeles, California, USA.,Doheny Eye Centers, Department of Ophthalmology, David Geffen School of Medicine at University of California, Los Angeles, California, USA
| | - Robert N Weinreb
- Shiley Eye Institute and Hamilton Glaucoma Center, Department of Ophthalmology, University of California, San Diego, California, USA
| |
Collapse
|
35
|
O'Callaghan J, Cassidy PS, Humphries P. Open-angle glaucoma: therapeutically targeting the extracellular matrix of the conventional outflow pathway. Expert Opin Ther Targets 2017; 21:1037-1050. [PMID: 28952395 DOI: 10.1080/14728222.2017.1386174] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
INTRODUCTION Ocular hypertension in open-angle glaucoma is caused by a reduced rate of removal of aqueous humour (AH) from the eye, with the majority of AH draining from the anterior chamber through the conventional outflow pathway, comprising the trabecular meshwork (TM) and Schlemm's Canal. Resistance to outflow is generated, in part, by the extracellular matrix (ECM) of the outflow tissues. Current pressure-lowering topical medications largely suppress AH production, or enhance its clearance through the unconventional pathway. However, therapies targeting the ECM of the conventional pathway in order to decrease intraocular pressure have become a recent focus of attention. Areas covered: We discuss the role of ECM of the TM in outflow homeostasis and its relevance as a target for glaucoma therapy, including progress in development of topical eye formulations, together with gene therapy approaches based on inducible, virally-mediated expression of matrix metalloproteinases to enhance aqueous outflow. Expert opinion: There remains a need for improved glaucoma medications that more specifically act upon sites causative to glaucoma pathogenesis. Emerging strategies targeting the ECM of the conventional outflow pathway, or associated components of the cytoskeleton of TM cells, involving new pharmacological formulations or genetically-based therapies, are promising avenues of future glaucoma treatment.
Collapse
Affiliation(s)
- Jeffrey O'Callaghan
- a Ocular Genetics Unit, Smurfit Institute of Genetics , University of Dublin, Trinity College , Dublin , Ireland
| | - Paul S Cassidy
- a Ocular Genetics Unit, Smurfit Institute of Genetics , University of Dublin, Trinity College , Dublin , Ireland
| | - Pete Humphries
- a Ocular Genetics Unit, Smurfit Institute of Genetics , University of Dublin, Trinity College , Dublin , Ireland
| |
Collapse
|
36
|
Liu H, Ding C. Establishment of an experimental glaucoma animal model: A comparison of microbead injection with or without hydroxypropyl methylcellulose. Exp Ther Med 2017; 14:1953-1960. [PMID: 28962109 PMCID: PMC5609141 DOI: 10.3892/etm.2017.4728] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 03/31/2017] [Indexed: 01/04/2023] Open
Abstract
The present study aimed to compare microbead injection with and without hydroxypropyl methylcellulose (HPM) in order to establish an experimental animal model of glaucoma. This model was established in C57BL/6 mice and transgenic mice expressing cyan fluorescent protein (CFP) under the control of the Thy1 promoter in retinal ganglion cells (RGCs). C57BL/6 mice aged between 12 and 20 weeks old were randomly separated into three groups, which received different injections into the anterior chamber of the eye. Group A (microbead) received 2 µl microbeads (10×106 beads/ml) and 1 µl air. Group B (microbeads + HPM) received 2 µl microbeads and 1 µl HPM. Group C (control group) received 2 µl PBS and 1 µl air. The intraocular pressure (IOP) was measured with a tonometer under topical anesthesia daily for 1 month. A single injection of microbeads, with or without HPM, induced consistent IOP elevation when compared with the control group. Thy1-CFP mice received an injection of 2 µl microbeads and 1 µl HPM into the anterior chamber of the eyes, and the number of CFP+ RGCs was subsequently assessed in vivo by confocal scanning laser microscopy in the same area of the retina weekly for 6 weeks. The results from in vivo imaging of Thy1-CFP mice were comparable with the immunohistochemical staining results from the C57BL/6 mice. The combined injection of microbeads and HPM induced longer and higher peaks of IOP elevation when compared with the microbeads alone. The rate of RGC loss following the administration of microbeads alone was 25.0±1.3% 6 weeks after the initial IOP elevation, while it was 33.2±1.9% following the administration of microbeads + HPM. These results indicate that the injection of microbeads + HPM is a more effective method of establishing a mouse model with chronic elevation of IOP. In addition, the in vivo imaging that can be used with this technique provides an effective and noninvasive approach for monitoring the progress of RGC loss.
Collapse
Affiliation(s)
- Hanhan Liu
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Chun Ding
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| |
Collapse
|
37
|
Huang AS, Saraswathy S, Dastiridou A, Begian A, Mohindroo C, Tan JCH, Francis BA, Hinton DR, Weinreb RN. Aqueous Angiography-Mediated Guidance of Trabecular Bypass Improves Angiographic Outflow in Human Enucleated Eyes. Invest Ophthalmol Vis Sci 2017; 57:4558-65. [PMID: 27588614 PMCID: PMC5017267 DOI: 10.1167/iovs.16-19644] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Purpose To assess the ability of trabecular micro-bypass stents to improve aqueous humor outflow (AHO) in regions initially devoid of AHO as assessed by aqueous angiography. Methods Enucleated human eyes (14 total from 7 males and 3 females [ages 52–84]) were obtained from an eye bank within 48 hours of death. Eyes were oriented by inferior oblique insertion, and aqueous angiography was performed with indocyanine green (ICG; 0.4%) or fluorescein (2.5%) at 10 mm Hg. With an angiographer, infrared and fluorescent images were acquired. Concurrent anterior segment optical coherence tomography (OCT) was performed, and fixable fluorescent dextrans were introduced into the eye for histologic analysis of angiographically positive and negative areas. Experimentally, some eyes (n = 11) first received ICG aqueous angiography to determine angiographic patterns. These eyes then underwent trabecular micro-bypass sham or stent placement in regions initially devoid of angiographic signal. This was followed by fluorescein aqueous angiography to query the effects. Results Aqueous angiography in human eyes yielded high-quality images with segmental patterns. Distally, angiographically positive but not negative areas demonstrated intrascleral lumens on OCT images. Aqueous angiography with fluorescent dextrans led to their trapping in AHO pathways. Trabecular bypass but not sham in regions initially devoid of ICG aqueous angiography led to increased aqueous angiography as assessed by fluorescein (P = 0.043). Conclusions Using sequential aqueous angiography in an enucleated human eye model system, regions initially without angiographic flow or signal could be recruited for AHO using a trabecular bypass stent.
Collapse
Affiliation(s)
- Alex S Huang
- Doheny Eye Institute, Los Angeles, California, United States 2Department of Ophthalmology, David Geffen School of Medicine at University of California-Los Angeles, Los Angeles, California, United States
| | | | - Anna Dastiridou
- Doheny Eye Institute, Los Angeles, California, United States
| | - Alan Begian
- Department of Ophthalmology, David Geffen School of Medicine at University of California-Los Angeles, Los Angeles, California, United States
| | | | - James C H Tan
- Doheny Eye Institute, Los Angeles, California, United States 2Department of Ophthalmology, David Geffen School of Medicine at University of California-Los Angeles, Los Angeles, California, United States
| | - Brian A Francis
- Doheny Eye Institute, Los Angeles, California, United States 2Department of Ophthalmology, David Geffen School of Medicine at University of California-Los Angeles, Los Angeles, California, United States
| | - David R Hinton
- Department of Ophthalmology and Pathology, University of Southern California, Los Angeles, California, United States
| | - Robert N Weinreb
- Hamilton Glaucoma Center and Shiley Eye Institute, University of California-San Diego, San Diego, California, United States
| |
Collapse
|
38
|
Ren R, Li G, Le TD, Kopczynski C, Stamer WD, Gong H. Netarsudil Increases Outflow Facility in Human Eyes Through Multiple Mechanisms. Invest Ophthalmol Vis Sci 2017; 57:6197-6209. [PMID: 27842161 PMCID: PMC5114035 DOI: 10.1167/iovs.16-20189] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Purpose Netarsudil is a Rho kinase/norepinephrine transporter inhibitor currently in phase 3 clinical development for glaucoma treatment. We investigated the effects of its active metabolite, netarsudil-M1, on outflow facility (C), outflow hydrodynamics, and morphology of the conventional outflow pathway in enucleated human eyes. Methods Paired human eyes (n = 5) were perfused with either 0.3 μM netarsudil-M1 or vehicle solution at constant pressure (15 mm Hg). After 3 hours, fluorescent microspheres were added to perfusion media to trace the outflow patterns before perfusion-fixation. The percentage effective filtration length (PEFL) was calculated from the measured lengths of tracer distribution in the trabecular meshwork (TM), episcleral veins (ESVs), and along the inner wall (IW) of Schlemm's canal after global and confocal imaging. Morphologic changes along the trabecular outflow pathway were investigated by confocal, light, and electron microscopy. Results Perfusion with netarsudil-M1 significantly increased C when compared to baseline (51%, P < 0.01) and to paired controls (102%, P < 0.01), as well as significantly increased PEFL in both IW (P < 0.05) and ESVs (P < 0.01). In treated eyes, PEFL was significantly higher in ESVs than in the IW (P < 0.01) and was associated with increased cross-sectional area of ESVs (P < 0.01). Percentage effective filtration length in ESVs positively correlated with the percentage change in C (R2 = 0.58, P = 0.01). A significant increase in juxtacanalicular connective tissue (JCT) thickness (P < 0.05) was found in treated eyes compared to controls. Conclusions Netarsudil acutely increased C by expansion of the JCT and dilating the ESVs, which led to redistribution of aqueous outflow through a larger area of the IW and ESVs.
Collapse
Affiliation(s)
- Ruiyi Ren
- Department of Ophthalmology, Boston University School of Medicine, Boston, Massachusetts, United States 2Department of Anatomoy and Neurobiology, Boston University School of Medicine, Boston, Massachusetts, United States
| | - Guorong Li
- Department of Ophthalmology, Duke University, Durham, North Carolina, United States
| | - Thuy Duong Le
- Department of Ophthalmology, Boston University School of Medicine, Boston, Massachusetts, United States
| | - Casey Kopczynski
- Aerie Pharmaceuticals, Inc., Durham, North Carolina, United States
| | - W Daniel Stamer
- Department of Ophthalmology, Duke University, Durham, North Carolina, United States
| | - Haiyan Gong
- Department of Ophthalmology, Boston University School of Medicine, Boston, Massachusetts, United States 2Department of Anatomoy and Neurobiology, Boston University School of Medicine, Boston, Massachusetts, United States
| |
Collapse
|
39
|
O'Callaghan J, Crosbie DE, Cassidy PS, Sherwood JM, Flügel-Koch C, Lütjen-Drecoll E, Humphries MM, Reina-Torres E, Wallace D, Kiang AS, Campbell M, Stamer WD, Overby DR, O'Brien C, Tam LCS, Humphries P. Therapeutic potential of AAV-mediated MMP-3 secretion from corneal endothelium in treating glaucoma. Hum Mol Genet 2017; 26:1230-1246. [PMID: 28158775 PMCID: PMC5390678 DOI: 10.1093/hmg/ddx028] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 01/18/2017] [Indexed: 11/13/2022] Open
Abstract
Intraocular pressure (IOP) is maintained as a result of the balance between production of aqueous humour (AH) by the ciliary processes and hydrodynamic resistance to its outflow through the conventional outflow pathway comprising the trabecular meshwork (TM) and Schlemm's canal (SC). Elevated IOP, which can be caused by increased resistance to AH outflow, is a major risk factor for open-angle glaucoma. Matrix metalloproteinases (MMPs) contribute to conventional aqueous outflow homeostasis in their capacity to remodel extracellular matrices, which has a direct impact on aqueous outflow resistance and IOP. We observed decreased MMP-3 activity in human glaucomatous AH compared to age-matched normotensive control AH. Treatment with glaucomatous AH resulted in significantly increased transendothelial resistance of SC endothelial and TM cell monolayers and reduced monolayer permeability when compared to control AH, or supplemented treatment with exogenous MMP-3.Intracameral inoculation of AAV-2/9 containing a CMV-driven MMP-3 gene (AAV-MMP-3) into wild type mice resulted in efficient transduction of corneal endothelium and an increase in aqueous concentration and activity of MMP-3. Most importantly, AAV-mediated expression of MMP-3 increased outflow facility and decreased IOP, and controlled expression using an inducible promoter activated by topical administration of doxycycline achieved the same effect. Ultrastructural analysis of MMP-3 treated matrices by transmission electron microscopy revealed remodelling and degradation of core extracellular matrix components. These results indicate that periodic induction, via use of an eye drop, of AAV-mediated secretion of MMP-3 into AH could have therapeutic potential for those cases of glaucoma that are sub-optimally responsive to conventional pressure-reducing medications.
Collapse
Affiliation(s)
- Jeffrey O'Callaghan
- Ocular Genetics Unit, Smurfit Institute of Genetics, University of Dublin, Trinity College, Dublin, D2, Ireland
| | - Darragh E Crosbie
- Ocular Genetics Unit, Smurfit Institute of Genetics, University of Dublin, Trinity College, Dublin, D2, Ireland
| | - Paul S Cassidy
- Ocular Genetics Unit, Smurfit Institute of Genetics, University of Dublin, Trinity College, Dublin, D2, Ireland
| | - Joseph M Sherwood
- Department of Bioengineering, Imperial College London, London, SW7 2BX, UK
| | - Cassandra Flügel-Koch
- Department of Anatomy II, University of Erlangen-Nürnberg, D-91054 Erlangen, Germany
| | - Elke Lütjen-Drecoll
- Department of Anatomy II, University of Erlangen-Nürnberg, D-91054 Erlangen, Germany
| | - Marian M Humphries
- Ocular Genetics Unit, Smurfit Institute of Genetics, University of Dublin, Trinity College, Dublin, D2, Ireland
| | - Ester Reina-Torres
- Ocular Genetics Unit, Smurfit Institute of Genetics, University of Dublin, Trinity College, Dublin, D2, Ireland
| | - Deborah Wallace
- Clinical Research Centre, UCD School of Medicine and Medical Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - Anna-Sophia Kiang
- Ocular Genetics Unit, Smurfit Institute of Genetics, University of Dublin, Trinity College, Dublin, D2, Ireland
| | - Matthew Campbell
- Ocular Genetics Unit, Smurfit Institute of Genetics, University of Dublin, Trinity College, Dublin, D2, Ireland
| | - W Daniel Stamer
- Departments of Ophthalmology and Biomedical Engineering, Duke University, Durham, NC, USA
| | - Darryl R Overby
- Department of Bioengineering, Imperial College London, London, SW7 2BX, UK
| | - Colm O'Brien
- Department of Ophthalmology, Mater Misericordiae University Hospital, Dublin, D7, Ireland
| | - Lawrence C S Tam
- Ocular Genetics Unit, Smurfit Institute of Genetics, University of Dublin, Trinity College, Dublin, D2, Ireland
| | - Peter Humphries
- Ocular Genetics Unit, Smurfit Institute of Genetics, University of Dublin, Trinity College, Dublin, D2, Ireland
| |
Collapse
|
40
|
Andrés-Guerrero V, García-Feijoo J, Konstas AG. Targeting Schlemm's Canal in the Medical Therapy of Glaucoma: Current and Future Considerations. Adv Ther 2017; 34:1049-1069. [PMID: 28349508 PMCID: PMC5427152 DOI: 10.1007/s12325-017-0513-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Indexed: 11/23/2022]
Abstract
Schlemm’s canal (SC) is a unique, complex vascular structure responsible for maintaining fluid homeostasis within the anterior segment of the eye by draining the excess of aqueous humour. In glaucoma, a heterogeneous group of eye disorders afflicting approximately 60 million individuals worldwide, the normal outflow of aqueous humour into SC is progressively hindered, leading to a gradual increase in outflow resistance, which gradually results in elevated intraocular pressure (IOP). By and large available antiglaucoma therapies do not target the site of the pathology (SC), but rather aim to decrease IOP by other mechanisms, either reducing aqueous production or by diverting aqueous flow through the unconventional outflow system. The present review first outlines our current understanding on the functional anatomy of SC. It then summarizes existing research on SC cell properties; first in the context of their role in glaucoma development/progression and then as a target of novel and emerging antiglaucoma therapies. Evidence from ongoing research efforts to develop effective antiglaucoma therapies targeting SC suggests that this could become a promising site of future therapeutic interventions.
Collapse
|
41
|
Carreon TA, Edwards G, Wang H, Bhattacharya SK. Segmental outflow of aqueous humor in mouse and human. Exp Eye Res 2017; 158:59-66. [PMID: 27498226 PMCID: PMC5290258 DOI: 10.1016/j.exer.2016.08.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Revised: 07/08/2016] [Accepted: 08/01/2016] [Indexed: 12/28/2022]
Abstract
The main and only modifiable risk factor in glaucoma, the group of usually late onset progressive and irreversible blinding optic neuropathies, is elevated intraocular pressure (IOP). The increase in IOP is due to impeded aqueous humor (AH) outflow through the conventional pathway. The aberrant increased resistance at the trabecular meshwork (TM), the filter-like region in the anterior eye chamber is the major contributory factor in causing the impeded outflow. In normal as well as in glaucoma eyes the regions of the TM are divided into areas of high and low flow. The collector channels and distal outflow regions are now increasingly being recognized as potential players in contributing to impede AH outflow. Structural and molecular make-up contributing to the segmental blockage to outflow is likely to provide greater insight. Establishing segmental blockage to outflow in model systems of glaucoma such as the mouse in parallel to human eyes will expand our repertoire of tools for investigation. Further study into this area of interest has the potential to ultimately lead to the development of new therapeutics focused on lowering IOP by targeting the various components of segmental blockage of outflow in the TM and in the distal outflow region.
Collapse
Affiliation(s)
- Teresia A Carreon
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA; Department of Biochemistry and Molecular Biology, University of Miami, Miami, FL, 33136, USA
| | - Genea Edwards
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA; Department of Biochemistry and Molecular Biology, University of Miami, Miami, FL, 33136, USA
| | - Haiyan Wang
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA; Shanghai First People's Hospital Affiliated to Jiaotong University, Shanghai, 200080, China
| | - Sanjoy K Bhattacharya
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA; Department of Biochemistry and Molecular Biology, University of Miami, Miami, FL, 33136, USA.
| |
Collapse
|
42
|
Carreon T, van der Merwe E, Fellman RL, Johnstone M, Bhattacharya SK. Aqueous outflow - A continuum from trabecular meshwork to episcleral veins. Prog Retin Eye Res 2017; 57:108-133. [PMID: 28028002 PMCID: PMC5350024 DOI: 10.1016/j.preteyeres.2016.12.004] [Citation(s) in RCA: 200] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 11/14/2016] [Accepted: 12/22/2016] [Indexed: 12/22/2022]
Abstract
In glaucoma, lowered intraocular pressure (IOP) confers neuroprotection. Elevated IOP characterizes glaucoma and arises from impaired aqueous humor (AH) outflow. Increased resistance in the trabecular meshwork (TM), a filter-like structure essential to regulate AH outflow, may result in the impaired outflow. Flow through the 360° circumference of TM structures may be non-uniform, divided into high and low flow regions, termed as segmental. After flowing through the TM, AH enters Schlemm's canal (SC), which expresses both blood and lymphatic markers; AH then passes into collector channel entrances (CCE) along the SC external well. From the CCE, AH enters a deep scleral plexus (DSP) of vessels that typically run parallel to SC. From the DSP, intrascleral collector vessels run radially to the scleral surface to connect with AH containing vessels called aqueous veins to discharge AH to blood-containing episcleral veins. However, the molecular mechanisms that maintain homeostatic properties of endothelial cells along the pathways are not well understood. How these molecular events change during aging and in glaucoma pathology remain unresolved. In this review, we propose mechanistic possibilities to explain the continuum of AH outflow control, which originates at the TM and extends through collector channels to the episcleral veins.
Collapse
Affiliation(s)
- Teresia Carreon
- Department of Ophthalmology & Bascom Palmer Eye Institute, University of Miami, Miami, USA; Department of Biochemistry and Molecular Biology, University of Miami, Miami, USA
| | - Elizabeth van der Merwe
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, 7925 Cape Town, South Africa
| | | | - Murray Johnstone
- Department of Ophthalmology, University of Washington, Seattle, WA, USA
| | - Sanjoy K Bhattacharya
- Department of Ophthalmology & Bascom Palmer Eye Institute, University of Miami, Miami, USA; Department of Biochemistry and Molecular Biology, University of Miami, Miami, USA.
| |
Collapse
|
43
|
Manabe SI, Sawaguchi S, Hayashi K. The effect of the extent of the incision in the Schlemm canal on the surgical outcomes of suture trabeculotomy for open-angle glaucoma. Jpn J Ophthalmol 2016; 61:99-104. [PMID: 27848108 DOI: 10.1007/s10384-016-0487-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Accepted: 09/25/2016] [Indexed: 11/26/2022]
Abstract
PURPOSE To examine the relationship between the extent of the incision in the Schlemm canal during suture trabeculotomy (S-LOT) for open-angle glaucoma and the associated reduction in intraocular pressure (IOP). METHODS Forty-eight consecutive eyes (primary open-angle glaucoma: 28; exfoliation glaucoma: 20) scheduled to undergo S-LOT were studied. A 320-degree opening in the Schlemm canal was planned when total circumferential cannulation had been achieved, and the remaining 40-degree region under the scleral flap was not incised. When the cannulation was incomplete, the canal was incised up to the point reached by the cannula, and the extent of the incision was measured. Logistic regression analysis was performed to identify potential predictors of surgical success (IOP ≤ 15 mmHg). RESULTS A 320-degree incision was achieved in 23 eyes, and the mean extent of the incision was 275 ± 52.3°. The mean IOP fell from 34.0 ± 8.8 mmHg at baseline to 14.5 ± 4.0 mmHg at 1 year postoperatively (P < 0.0001). An IOP reduction of ≥30% was achieved in 46 eyes. Simple correlation analysis indicated that the extent of the incision in degrees did not correlate with IOP or with the reduction in IOP at 1 year postoperatively (P = 0.7665 and 0.693, respectively). Lower preoperative IOP values and concomitant cataract extraction were found to be associated with surgical success. CONCLUSIONS The extent of the incision in the Schlemm canal during S-LOT does not correlate with postoperative reduction in IOP.
Collapse
Affiliation(s)
- Shin-Ichi Manabe
- Hayashi Eye Hospital, 4-23-35 Hakataekimae, Hakata-ku, Fukuoka, 812-0011, Japan.
| | - Shoichi Sawaguchi
- Department of Ophthalmology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Ken Hayashi
- Hayashi Eye Hospital, 4-23-35 Hakataekimae, Hakata-ku, Fukuoka, 812-0011, Japan
| |
Collapse
|
44
|
Huang AS, Saraswathy S, Dastiridou A, Begian A, Legaspi H, Mohindroo C, Tan JCH, Francis BA, Caprioli J, Hinton DR, Weinreb RN. Aqueous Angiography with Fluorescein and Indocyanine Green in Bovine Eyes. Transl Vis Sci Technol 2016; 5:5. [PMID: 27847692 PMCID: PMC5106193 DOI: 10.1167/tvst.5.6.5] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 09/13/2016] [Indexed: 11/28/2022] Open
Abstract
Purpose We characterize aqueous angiography as a real-time aqueous humor outflow imaging (AHO) modality in cow eyes with two tracers of different molecular characteristics. Methods Cow enucleated eyes (n = 31) were obtained and perfused with balanced salt solution via a Lewicky AC maintainer through a 1-mm side-port. Fluorescein (2.5%) or indocyanine green (ICG; 0.4%) were introduced intracamerally at 10 mm Hg individually or sequentially. With an angiographer, infrared and fluorescent images were acquired. Concurrent anterior segment optical coherence tomography (OCT) was performed, and fixable fluorescent dextrans were introduced into the eye for histologic analysis of angiographically positive and negative areas. Results Aqueous angiography in cow eyes with fluorescein and ICG yielded high-quality images with segmental patterns. Over time, ICG maintained a better intraluminal presence. Angiographically positive, but not negative, areas demonstrated intrascleral lumens with anterior segment OCT. Aqueous angiography with fluorescent dextrans led to their trapping in AHO pathways. Sequential aqueous angiography with ICG followed by fluorescein in cow eyes demonstrated similar patterns. Conclusions Aqueous angiography in model cow eyes demonstrated segmental angiographic outflow patterns with either fluorescein or ICG as a tracer. Translational Relevance Further characterization of segmental AHO with aqueous angiography may allow for intelligent placement of trabecular bypass minimally invasive glaucoma surgeries for improved surgical results.
Collapse
Affiliation(s)
- Alex S Huang
- Doheny Eye Institute, Los Angeles, CA, USA ; Department of Ophthalmology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | | | | | - Alan Begian
- Doheny Eye Institute, Los Angeles, CA, USA ; Department of Ophthalmology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Hanz Legaspi
- Doheny Eye Institute, Los Angeles, CA, USA ; Department of Ophthalmology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | | | - James C H Tan
- Doheny Eye Institute, Los Angeles, CA, USA ; Department of Ophthalmology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Brian A Francis
- Doheny Eye Institute, Los Angeles, CA, USA ; Department of Ophthalmology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Joseph Caprioli
- Department of Ophthalmology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA ; Stein Eye Institute, Los Angeles, CA, USA
| | - David R Hinton
- Department of Ophthalmology and Pathology, University of Southern California, Los Angeles, CA, USA
| | - Robert N Weinreb
- Hamilton Glaucoma Center and Shiley Eye Institute, University of California, San Diego, CA, USA
| |
Collapse
|
45
|
Huang AS, Mohindroo C, Weinreb RN. Aqueous Humor Outflow Structure and Function Imaging At the Bench and Bedside: A Review. ACTA ACUST UNITED AC 2016; 7. [PMID: 27790380 PMCID: PMC5079182 DOI: 10.4172/2155-9570.1000578] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
Anterior segment glaucoma clinical care and research has recently gained new focus because of novel imaging modalities and the advent of angle-based surgical treatments. Traditional investigation drawn to the trabecular meshwork now emphasizes the entire conventional aqueous humor outflow (AHO) pathway from the anterior chamber to the episcleral vein. AHO investigation can be divided into structural and functional assessments using different methods. The historical basis for studying the anterior segment of the eye and AHO in glaucoma is discussed. Structural studies of AHO are reviewed and include traditional pathological approaches to modern tools such as multi-model two-photon microscopy and optical coherence tomography. Functional assessment focuses on visualizing AHO itself through a variety of non-real-time and real-time techniques such as aqueous angiography. Implications of distal outflow resistance and segmental AHO are discussed with an emphasis on melding bench-side research to viable clinical applications. Through the development of an improved structure: function relationship for AHO in the anterior segment of the normal and diseased eye, a better understanding of the eye with improved therapeutics may be developed.
Collapse
Affiliation(s)
- Alex S Huang
- Doheny Eye Institute, Los Angeles, CA, USA; Department of Ophthalmology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | | | - Robert N Weinreb
- Hamilton Glaucoma Center and Shiley Eye Institute, University of California, San Diego, CA, USA
| |
Collapse
|
46
|
Trabecular meshwork stiffness in glaucoma. Exp Eye Res 2016; 158:3-12. [PMID: 27448987 DOI: 10.1016/j.exer.2016.07.011] [Citation(s) in RCA: 113] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Revised: 07/15/2016] [Accepted: 07/18/2016] [Indexed: 12/26/2022]
Abstract
Alterations in stiffness of the trabecular meshwork (TM) may play an important role in primary open-angle glaucoma (POAG), the second leading cause of blindness. Specifically, certain data suggest an association between elevated intraocular pressure (IOP) and increased TM stiffness; however, the underlying link between TM stiffness and IOP remains unclear and requires further study. We here first review the literature on TM stiffness measurements, encompassing various species and based on a number of measurement techniques, including direct approaches such as atomic force microscopy (AFM) and uniaxial tension tests, and indirect methods based on a beam deflection model. We also briefly review the effects of several factors that affect TM stiffness, including lysophospholipids, rho-kinase inhibitors, cytoskeletal disrupting agents, dexamethasone (DEX), transforming growth factor-β2 (TGF-β2), nitric oxide (NO) and cellular senescence. We then describe a method we have developed for determining TM stiffness measurement in mice using a cryosection/AFM-based approach, and present preliminary data on TM stiffness in C57BL/6J and CBA/J mouse strains. Finally, we investigate the relationship between TM stiffness and outflow facility between these two strains. The method we have developed shows promise for further direct measurements of mouse TM stiffness, which may be of value in understanding mechanistic relations between outflow facility and TM biomechanical properties.
Collapse
|
47
|
Vranka JA, Acott TS. Pressure-induced expression changes in segmental flow regions of the human trabecular meshwork. Exp Eye Res 2016; 158:67-72. [PMID: 27334250 DOI: 10.1016/j.exer.2016.06.009] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 06/11/2016] [Accepted: 06/16/2016] [Indexed: 10/24/2022]
Abstract
Elevated intraocular pressure (IOP) is thought to create distortion or stretching of the juxtacanalicular and Schlemm's canal cells and their extracellular matrix (ECM) leading to a cascade of events that restore IOP to normal levels, a process termed IOP homeostasis. The ECM of the trabecular meshwork (TM) is intricately involved in the regulation of outflow resistance and IOP homeostasis, as matrix metalloproteinase (MMP)-initiated ECM turnover in the TM is necessary to maintain outflow facility. Previous studies have shown ECM gene expression and mRNA splice form differences in TM cells in response to sustained stretch, implicating their involvement in the dynamic process of IOP homeostasis. The observation that outflow is segmental around the circumference of the eye adds another layer of complexity to understanding the molecular events necessary to maintaining proper outflow facility. The aim of this work was to identify molecular expression differences between segmental flow regions of the TM from anterior segments perfused at either physiological or elevated pressure. Human anterior segments were perfused in an ex vivo model system, TM tissues were extracted and quantitative PCR arrays were performed. Comparisons were made between high flow and low flow regions of the TM from anterior segments perfused either at normal (8.8 mmHg) or at elevated (17.6 mmHg) perfusion pressure for 48 h. The results are presented here as independent sets: 1) fold change gene expression between segmental flow regions at a single perfusion pressure, and 2) fold change gene expression in response to elevated perfusion pressure in a single flow region. Multiple genes from the following functional families were found to be differentially expressed in segmental regions and in response to elevated pressure: collagens, ECM glycoproteins including matricellular proteins, ECM receptors such as integrins and adhesion molecules and ECM regulators, such as matrix metalloproteinases. In general, under normal perfusion pressure, more ECM genes were enriched in the high flow regions than in the low flow regions of the TM, whereas more ECM genes were found to be enriched in low flow regions of the TM in response to elevated perfusion pressure. Thus it appears that a limited subset of ECM genes is differentially regulated in both high and low flow regions and in response to elevated pressure. Some of these same ECM genes have previously been shown to be involved in the pressure response of stretched TM cells supporting their central role in IOP homeostasis. In general, different ECM gene family members are called upon to produce the response to elevated pressure in different segmental regions of the TM.
Collapse
Affiliation(s)
- Janice A Vranka
- Casey Eye Institute, Oregon Health & Science University, Portland, OR, 97239, USA.
| | - Ted S Acott
- Casey Eye Institute, Oregon Health & Science University, Portland, OR, 97239, USA
| |
Collapse
|
48
|
The exit strategy: Pharmacological modulation of extracellular matrix production and deposition for better aqueous humor drainage. Eur J Pharmacol 2016; 787:32-42. [PMID: 27112663 DOI: 10.1016/j.ejphar.2016.04.048] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 04/20/2016] [Accepted: 04/22/2016] [Indexed: 01/28/2023]
Abstract
Primary open angle glaucoma (POAG) is an optic neuropathy and an irreversible blinding disease. The etiology of glaucoma is not known but numerous risk factors are associated with this disease including aging, elevated intraocular pressure (IOP), race, myopia, family history and use of steroids. In POAG, the resistance to the aqueous humor drainage is increased leading to elevated IOP. Lowering the resistance and ultimately the IOP has been the only way to slow disease progression and prevent vision loss. The primary drainage pathway comprising of the trabecular meshwork (TM) is made up of relatively large porous beams surrounded by extracellular matrix (ECM). Its juxtacanalicular tissue (JCT) or the cribriform meshwork is made up of cells embedded in dense ECM. The JCT is considered to offer the major resistance to the aqueous humor outflow. This layer is adjacent to the endothelial cells forming Schlemm's canal, which provides approximately 10% of the outflow resistance. The ECM in the TM and the JCT undergoes continual remodeling to maintain normal resistance to aqueous humor outflow. It is believed that the TM is a major contributor of ECM proteins and evidence points towards increased ECM deposition in the outflow pathway in POAG. It is not clear how and from where the ECM components emerge to hinder the normal aqueous humor drainage. This review focuses on the involvement of the ECM in ocular hypertension and glaucoma and the mechanisms by which various ocular hypotensive drugs, both current and emerging, target ECM production, remodeling, and deposition.
Collapse
|
49
|
Li G, Mukherjee D, Navarro I, Ashpole NE, Sherwood JM, Chang J, Overby DR, Yuan F, Gonzalez P, Kopczynski CC, Farsiu S, Stamer WD. Visualization of conventional outflow tissue responses to netarsudil in living mouse eyes. Eur J Pharmacol 2016; 787:20-31. [PMID: 27085895 DOI: 10.1016/j.ejphar.2016.04.002] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 03/10/2016] [Accepted: 04/04/2016] [Indexed: 12/23/2022]
Abstract
Visual impairment due to glaucoma currently impacts 70 million people worldwide. While disease progression can be slowed or stopped with effective lowering of intraocular pressure, current medical treatments are often inadequate. Fortunately, three new classes of therapeutics that target the diseased conventional outflow tissue responsible for ocular hypertension are in the final stages of human testing. The rho kinase inhibitors have proven particularly efficacious and additive to current therapies. Unfortunately, non-contact technology that monitors the health of outflow tissue and its response to conventional outflow therapy is not available clinically. Using optical coherence tomographic (OCT) imaging and novel segmentation software, we present the first demonstration of drug effects on conventional outflow tissues in living eyes. Topical netarsudil (formerly AR-13324), a rho kinase/ norepinephrine transporter inhibitor, affected both proximal (trabecular meshwork and Schlemm's Canal) and distal portions (intrascleral vessels) of the mouse conventional outflow tract. Hence, increased perfusion of outflow tissues was reliably resolved by OCT as widening of the trabecular meshwork and significant increases in cross-sectional area of Schlemm's canal following netarsudil treatment. These changes occurred in conjunction with increased outflow facility, increased speckle variance intensity of outflow vessels, increased tracer deposition in conventional outflow tissues and decreased intraocular pressure. This is the first report using live imaging to show real-time drug effects on conventional outflow tissues and specifically the mechanism of action of netarsudil in mouse eyes. Advancements here pave the way for development of a clinic-friendly OCT platform for monitoring glaucoma therapy.
Collapse
Affiliation(s)
- Guorong Li
- Department of Ophthalmology, Duke University, Durham, NC 27710, USA
| | - Dibyendu Mukherjee
- Department of Biomedical Engineering, Duke University, Durham, NC 27710, USA
| | - Iris Navarro
- Department of Ophthalmology, Duke University, Durham, NC 27710, USA
| | - Nicole E Ashpole
- Department of Ophthalmology, Duke University, Durham, NC 27710, USA
| | - Joseph M Sherwood
- Department of Bioengineering, Imperial College London, London SW7 2AZ, United Kingdom
| | - Jinlong Chang
- Department of Biomedical Engineering, Duke University, Durham, NC 27710, USA
| | - Darryl R Overby
- Department of Bioengineering, Imperial College London, London SW7 2AZ, United Kingdom
| | - Fan Yuan
- Department of Biomedical Engineering, Duke University, Durham, NC 27710, USA
| | - Pedro Gonzalez
- Department of Ophthalmology, Duke University, Durham, NC 27710, USA
| | | | - Sina Farsiu
- Department of Ophthalmology, Duke University, Durham, NC 27710, USA; Department of Biomedical Engineering, Duke University, Durham, NC 27710, USA
| | - W Daniel Stamer
- Department of Ophthalmology, Duke University, Durham, NC 27710, USA; Department of Biomedical Engineering, Duke University, Durham, NC 27710, USA.
| |
Collapse
|
50
|
Aqueous Angiography: Real-Time and Physiologic Aqueous Humor Outflow Imaging. PLoS One 2016; 11:e0147176. [PMID: 26807586 PMCID: PMC4725949 DOI: 10.1371/journal.pone.0147176] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 12/30/2015] [Indexed: 01/22/2023] Open
Abstract
Purpose Trabecular meshwork (TM) bypass surgeries attempt to enhance aqueous humor outflow (AHO) to lower intraocular pressure (IOP). While TM bypass results are promising, inconsistent success is seen. One hypothesis for this variability rests upon segmental (non-360 degrees uniform) AHO. We describe aqueous angiography as a real-time and physiologic AHO imaging technique in model eyes as a way to simulate live AHO imaging. Methods Pig (n = 46) and human (n = 6) enucleated eyes were obtained, orientated based upon inferior oblique insertion, and pre-perfused with balanced salt solution via a Lewicky AC maintainer through a 1mm side-port. Fluorescein (2.5%) was introduced intracamerally at 10 or 30 mm Hg. With an angiographer, infrared and fluorescent (486 nm) images were acquired. Image processing allowed for collection of pixel information based on intensity or location for statistical analyses. Concurrent OCT was performed, and fixable fluorescent dextrans were introduced into the eye for histological analysis of angiographically active areas. Results Aqueous angiography yielded high quality images with segmental patterns (p<0.0001; Kruskal-Wallis test). No single quadrant was consistently identified as the primary quadrant of angiographic signal (p = 0.06–0.86; Kruskal-Wallis test). Regions of high proximal signal did not necessarily correlate with regions of high distal signal. Angiographically positive but not negative areas demonstrated intrascleral lumens on OCT images. Aqueous angiography with fluorescent dextrans led to their trapping in AHO pathways. Conclusions Aqueous angiography is a real-time and physiologic AHO imaging technique in model eyes.
Collapse
|