1
|
Asrani SG, McGlumphy EJ, Al-Aswad LA, Chaya CJ, Lin S, Musch DC, Pitha I, Robin AL, Wirostko B, Johnson TV. The relationship between intraocular pressure and glaucoma: An evolving concept. Prog Retin Eye Res 2024; 103:101303. [PMID: 39303763 DOI: 10.1016/j.preteyeres.2024.101303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 09/17/2024] [Accepted: 09/18/2024] [Indexed: 09/22/2024]
Abstract
Intraocular pressure (IOP) is the most important modifiable risk factor for glaucoma and fluctuates considerably within patients over short and long time periods. Our field's understanding of IOP has evolved considerably in recent years, driven by tonometric technologies with increasing accuracy, reproducibility, and temporal resolution that have refined our knowledge regarding the relationship between IOP and glaucoma risk and pathogenesis. The goal of this article is to review the published literature pertinent to the following points: 1) the factors that determine IOP in physiologic and pathologic states; 2) technologies for measuring IOP; 3) scientific and clinical rationale for measuring diverse IOP metrics in patients with glaucoma; 4) the impact and shortcomings of current standard-of-care IOP monitoring approaches; 5) recommendations for approaches to IOP monitoring that could improve patient outcomes; and 6) research questions that must be answered to improve our understanding of how IOP contributes to disease progression. Retrospective and prospective data, including that from landmark clinical trials, document greater IOP fluctuations in glaucomatous than healthy eyes, tendencies for maximal daily IOP to occur outside of office hours, and, in addition to mean and maximal IOP, an association between IOP fluctuation and glaucoma progression that is independent of mean in-office IOP. Ambulatory IOP monitoring, measuring IOP outside of office hours and at different times of day and night, provides clinicians with discrete data that could improve patient outcomes. Eye care clinicians treating glaucoma based on isolated in-office IOP measurements may make treatment decisions without fully capturing the entire IOP profile of an individual. Data linking home blood pressure monitors and home glucose sensors to dramatically improved outcomes for patients with systemic hypertension and diabetes and will be reviewed as they pertain to the question of whether ambulatory tonometry is positioned to do the same for glaucoma management. Prospective randomized controlled studies are warranted to determine whether remote tonometry-based glaucoma management might reduce vision loss and improve patient outcomes.
Collapse
Affiliation(s)
- Sanjay G Asrani
- Department of Ophthalmology, Duke University Medical Center, Durham, NC, USA
| | | | - Lama A Al-Aswad
- Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Craig J Chaya
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA
| | - Shan Lin
- Glaucoma Center of San Francisco, San Francisco, CA, USA
| | - David C Musch
- Department of Ophthalmology & Visual Sciences and Department of Epidemiology, Kellogg Eye Center, University of Michigan, Ann Arbor, MI, USA
| | - Ian Pitha
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alan L Robin
- Department of Ophthalmology & Visual Sciences and Department of Epidemiology, Kellogg Eye Center, University of Michigan, Ann Arbor, MI, USA; Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Barbara Wirostko
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA.
| | - Thomas V Johnson
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
2
|
Zemitis A, Vanags J, Fan J, Klavins K, Laganovska G. Metabolomic Disparities in Intraocular Fluid Across Varied Stages of Cataract Progression: Implications for the Analysis of Cataract Development. J Ocul Pharmacol Ther 2024; 40:477-485. [PMID: 38976556 DOI: 10.1089/jop.2024.0067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/10/2024] Open
Abstract
Introduction: The lens's metabolic demands are met through a continuous circulation of aqueous humor, encompassing a spectrum of components such as organic and inorganic ions, carbohydrates, glutathione, urea, amino acids, proteins, oxygen, carbon dioxide, and water. Metabolomics is a pivotal tool, offering an initial insight into the complexities of integrated metabolism. In this investigative study, we systematically scrutinize the composition of intraocular fluid in individuals afflicted with cataracts. Methods: The investigation involved a comprehensive analysis of aqueous humor samples from a cohort comprising 192 patients. These individuals were stratified by utilizing the SPONCS classification system, delineating distinct groups characterized by the hardness of cataracts. The analytical approach employed targeted quantitative metabolite analysis using HILIC-based liquid chromatography coupled with high-resolution mass spectrometric detection. The metabolomics data analysis was performed with MetaboAnalyst 5.0. Results: The results of the enrichment analysis have facilitated the inference that the discerned disparities among groups arise from disruptions in taurine and hypotaurine metabolism, variations in tryptophan metabolism, and modifications in mitochondrial beta-oxidation of short-chain saturated fatty acids and pyrimidine metabolism. Conclusion: A decline in taurine concentration precipitates diminished glutathione activity, prompting an elevated requirement for NAD+ and instigating tryptophan metabolism along the kynurenine pathway. Activation of this pathway is additionally prompted by interferon-gamma and UV radiation, leading to the induction of IDO. Concurrently, heightened mitochondrial beta-oxidation signifies a distinctive scenario in translocating fatty acids into the mitochondria, enhancing energy production.
Collapse
Affiliation(s)
- Arturs Zemitis
- Department of Ophthalmology, Riga Stradins University, Riga, Latvia
- Clinic of Ophthalmology, Pauls Stradins Clinical University Hospital, Riga, Latvia
| | - Juris Vanags
- Department of Ophthalmology, Riga Stradins University, Riga, Latvia
- Clinic of Ophthalmology, Pauls Stradins Clinical University Hospital, Riga, Latvia
| | - Jingzhi Fan
- Faculty of Natural Sciences and Technology, Institute of Biomaterials and Bioengineering, Riga Technical University, Riga, Latvia
| | - Kristaps Klavins
- Faculty of Natural Sciences and Technology, Institute of Biomaterials and Bioengineering, Riga Technical University, Riga, Latvia
| | - Guna Laganovska
- Department of Ophthalmology, Riga Stradins University, Riga, Latvia
- Clinic of Ophthalmology, Pauls Stradins Clinical University Hospital, Riga, Latvia
| |
Collapse
|
3
|
Benagiano V, Rizzi A, Sannace C, Alessio G, Ribatti D, Dammacco R. Aqueous humor as eye lymph: A crossroad between venous and lymphatic system. Exp Eye Res 2024; 243:109904. [PMID: 38642600 DOI: 10.1016/j.exer.2024.109904] [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: 12/15/2023] [Revised: 03/18/2024] [Accepted: 04/17/2024] [Indexed: 04/22/2024]
Abstract
Aqueous humor (AQH) is a transparent fluid with characteristics similar to those of the interstitial fluid, which fills the eyeball posterior and anterior chambers and circulates in them from the sites of production to those of drainage. The AQH volume and pressure homeostasis is essential for the trophism of the ocular avascular tissues and their normal structure and function. Different AQH outflow pathways exist, including a main pathway, quite well defined anatomically and referred to as the conventional pathway, and some accessory pathways, more recently described and still not fully morphofunctionally understood, generically referred to as unconventional pathways. The conventional pathway is based on the existence of a series of conduits starting with the trabecular meshwork and Schlemm's Canal and continuing with a system of intrascleral and episcleral venules, which are tributaries to veins of the anterior segment of the eyeball. The unconventional pathways are mainly represented by the uveoscleral pathway, in which AQH flows through clefts, interstitial conduits located in the ciliary body and sclera, and then merges into the aforementioned intrascleral and episcleral venules. A further unconventional pathway, the lymphatic pathway, has been supported by the demonstration of lymphatic microvessels in the limbal sclera and, possibly, in the uvea (ciliary body, choroid) as well as by the ocular glymphatic channels, present in the neural retina and optic nerve. It follows that AQH may be drained from the eyeball through blood vessels (TM-SC pathway, US pathway) or lymphatic vessels (lymphatic pathway), and the different pathways may integrate or compensate for each other, optimizing the AQH drainage. The present review aims to define the state-of-the-art concerning the structural organization and the functional anatomy of all the AQH outflow pathways. Particular attention is paid to examining the regulatory mechanisms active in each of them. The new data on the anatomy and physiology of AQH outflow pathways is the key to understanding the pathophysiology of AQH outflow disorders and could open the way for novel approaches to their treatment.
Collapse
Affiliation(s)
- Vincenzo Benagiano
- Department of Translational Biomedicine and Neuroscience, University of Bari 'Aldo Moro', Bari, Italy.
| | - Anna Rizzi
- Department of Translational Biomedicine and Neuroscience, University of Bari 'Aldo Moro', Bari, Italy
| | - Carmela Sannace
- Azienda Sanitaria Locale Bari, Ophthalmology Day Service Triggiano-Gioia del Colle, Bari, Italy
| | - Giovanni Alessio
- Department of Translational Biomedicine and Neuroscience, University of Bari 'Aldo Moro', Bari, Italy
| | - Domenico Ribatti
- Department of Translational Biomedicine and Neuroscience, University of Bari 'Aldo Moro', Bari, Italy
| | - Rosanna Dammacco
- Department of Translational Biomedicine and Neuroscience, University of Bari 'Aldo Moro', Bari, Italy
| |
Collapse
|
4
|
Kanjee R, Popovic MM, Salimi A, Hutnik CML, Ahmed IIK, Saheb H. Prophylaxis against intraocular pressure spikes following uncomplicated phacoemulsification: a systematic-review and meta-analysis. Eye (Lond) 2024; 38:1518-1528. [PMID: 38326483 PMCID: PMC11126686 DOI: 10.1038/s41433-024-02940-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 12/27/2023] [Accepted: 01/16/2024] [Indexed: 02/09/2024] Open
Abstract
BACKGROUND To investigate the effect of perioperative intraocular pressure (IOP) lowering medications on controlling postoperative IOP following uncomplicated phacoemulsification. METHODS Ovid MEDLINE, EMBASE, and Cochrane CENTRAL databases were searched up until November 2022. Randomised controlled trials (RCTs) that assessed IOP change via applanation tonometry in medicated and control arms following uncomplicated cataract surgery in healthy eyes were included. The primary outcome was the weighted mean difference (WMD) of IOP at 2-8 h, 12-24 h, and 1-7 days postoperatively within each medication class or common fixed-combination formulations. Risk of bias was assessed using the revised risk of bias in randomised trials (RoB-2). Level of evidence was rated using the Grading of Recommendation, Assessment, Development and Evaluation (GRADE) RESULTS: From 702 screened articles, 30 RCTs involving 2986 eyes were included. There was a statistically significant reduction in IOP favouring treatment arms at 2-8 h (WMD = -3.87 mmHg; 95% CI [-4.75, -3.00]; p < 0.001) and 12-24 h (WMD = -2.69 mmHg; 95% CI [-3.36, -2.02]; p < 0.001), with the effect wearing off beyond 1 day (p = 0.18). Between medication classes, the largest effect at both 2-8 h and 12-24 h was observed with intracameral cholinergics or fixed-combination carbonic anhydrase inhibitor-beta-blocker (FCCB) formulations. Conversely, the smallest effect was observed with prostaglandin analogues, alpha-agonists, and topical carbonic anhydrase inhibitors (CAIs). CONCLUSION Prophylaxis against acute IOP elevations following uncomplicated cataract surgery is effective. FCCB and intracameral cholinergics are the most effective ocular antihypertensive agents, while alpha-agonists, prostaglandin analogues, and topical CAIs were found to be the least effective. These findings may inform future surgical guidelines.
Collapse
Affiliation(s)
- Raageen Kanjee
- Department of Ophthalmology, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - Marko M Popovic
- Department of Ophthalmology and Vision Sciences, University of Toronto, Toronto, ON, Canada
| | - Ali Salimi
- Department of Ophthalmology & Visual Sciences, McGill University, Montreal, QC, Canada
| | | | - Iqbal Ike K Ahmed
- Department of Ophthalmology and Vision Sciences, University of Toronto, Toronto, ON, Canada
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA
| | - Hady Saheb
- Department of Ophthalmology & Visual Sciences, McGill University, Montreal, QC, Canada.
| |
Collapse
|
5
|
Barathi VA, Katz A, Chaudhary S, Li HL, Tal DM, Marcovich A, Do CW, Karlish SJD. A digoxin derivative that potently reduces intraocular pressure: efficacy and mechanism of action in different animal models. Am J Physiol Cell Physiol 2024; 326:C1505-C1519. [PMID: 38557355 PMCID: PMC11371363 DOI: 10.1152/ajpcell.00617.2023] [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: 11/15/2023] [Revised: 03/25/2024] [Accepted: 03/27/2024] [Indexed: 04/04/2024]
Abstract
Glaucoma is a blinding disease. Reduction of intraocular pressure (IOP) is the mainstay of treatment, but current drugs show side effects or become progressively ineffective, highlighting the need for novel compounds. We have synthesized a family of perhydro-1,4-oxazepine derivatives of digoxin, the selective inhibitor of Na,K-ATPase. The cyclobutyl derivative (DcB) displays strong selectivity for the human α2 isoform and potently reduces IOP in rabbits. These observations appeared consistent with a hypothesis that in ciliary epithelium DcB inhibits the α2 isoform of Na,K-ATPase, which is expressed strongly in nonpigmented cells, reducing aqueous humor (AH) inflow. This paper extends assessment of efficacy and mechanism of action of DcB using an ocular hypertensive nonhuman primate model (OHT-NHP) (Macaca fascicularis). In OHT-NHP, DcB potently lowers IOP, in both acute (24 h) and extended (7-10 days) settings, accompanied by increased aqueous humor flow rate (AFR). By contrast, ocular normotensive animals (ONT-NHP) are poorly responsive to DcB, if at all. The mechanism of action of DcB has been analyzed using isolated porcine ciliary epithelium and perfused enucleated eyes to study AH inflow and AH outflow facility, respectively. 1) DcB significantly stimulates AH inflow although prior addition of 8-Br-cAMP, which raises AH inflow, precludes additional effects of DcB. 2) DcB significantly increases AH outflow facility via the trabecular meshwork (TM). Taken together, the data indicate that the original hypothesis on the mechanism of action must be revised. In the OHT-NHP, and presumably other species, DcB lowers IOP by increasing AH outflow facility rather than by decreasing AH inflow.NEW & NOTEWORTHY When applied topically, a cyclobutyl derivative of digoxin (DcB) potently reduces intraocular pressure in an ocular hypertensive nonhuman primate model (Macaca fascicularis), associated with increased aqueous humor (AH) flow rate (AFR). The mechanism of action of DcB involves increased AH outflow facility as detected in enucleated perfused porcine eyes and, in parallel, increased (AH) inflow as detected in isolated porcine ciliary epithelium. DcB might have potential as a drug for the treatment of open-angle human glaucoma.
Collapse
Affiliation(s)
- Veluchamy Amutha Barathi
- Translational Pre-Clinical Model Platform, Singapore Institute of Eye Research (SERI)
- ACP in Ophthalmology & Visual Sciences, DUKE-NUS Graduate Medical School, Singapore
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Adriana Katz
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Shashikant Chaudhary
- School of Optometry, The Hong Kong Polytechnic University, Hong Kong, People's Republic of China
| | - Hoi-Lam Li
- School of Optometry, The Hong Kong Polytechnic University, Hong Kong, People's Republic of China
| | - Daniel M Tal
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Arie Marcovich
- Opthalmology Department, Kaplan Medical Center, Rehovot, Israel
- Hebrew University Medical School, Jerusalem, Israel
| | - Chi-Wai Do
- School of Optometry, The Hong Kong Polytechnic University, Hong Kong, People's Republic of China
- Centre for Eye and Vision Research (CEVR), Hong Kong, People's Republic of China
- Research Institute for Smart Ageing (RISA), The Hong Kong Polytechnic University, Hong Kong, People's Republic of China
| | - Steven J D Karlish
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
6
|
Wang CL, Skeie JM, Allamargot C, Goldstein AS, Nishimura DY, Huffman JM, Aldrich BT, Schmidt GA, Teixeira LBC, Kuehn MH, Yorek M, Greiner MA. Rat Model of Type 2 Diabetes Mellitus Recapitulates Human Disease in the Anterior Segment of the Eye. THE AMERICAN JOURNAL OF PATHOLOGY 2024:S0002-9440(24)00073-7. [PMID: 38403162 DOI: 10.1016/j.ajpath.2024.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 02/01/2024] [Accepted: 02/09/2024] [Indexed: 02/27/2024]
Abstract
Changes in the anterior segment of the eye due to type 2 diabetes mellitus (T2DM) are not well-characterized, in part due to the lack of a reliable animal model. This study evaluates changes in the anterior segment, including crystalline lens health, corneal endothelial cell density, aqueous humor metabolites, and ciliary body vasculature, in a rat model of T2DM compared with human eyes. Male Sprague-Dawley rats were fed a high-fat diet (45% fat) or normal diet, and rats fed the high-fat diet were injected with streptozotocin i.p. to generate a model of T2DM. Cataract formation and corneal endothelial cell density were assessed using microscopic analysis. Diabetes-related rat aqueous humor alterations were assessed using metabolomics screening. Transmission electron microscopy was used to assess qualitative ultrastructural changes ciliary process microvessels at the site of aqueous formation in the eyes of diabetic rats and humans. Eyes from the diabetic rats demonstrated cataracts, lower corneal endothelial cell densities, altered aqueous metabolites, and ciliary body ultrastructural changes, including vascular endothelial cell activation, pericyte degeneration, perivascular edema, and basement membrane reduplication. These findings recapitulated diabetic changes in human eyes. These results support the use of this model for studying ocular manifestations of T2DM and support a hypothesis postulating blood-aqueous barrier breakdown and vascular leakage at the ciliary body as a mechanism for diabetic anterior segment pathology.
Collapse
Affiliation(s)
- Cheryl L Wang
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Jessica M Skeie
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, Iowa; Iowa Lions Eye Bank, Coralville, Iowa
| | - Chantal Allamargot
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, Iowa; Office of the Vice President for Research, Central Microscopy Research Facility, University of Iowa, Iowa City, Iowa
| | - Andrew S Goldstein
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, Iowa; Iowa Lions Eye Bank, Coralville, Iowa
| | - Darryl Y Nishimura
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, Iowa; Iowa Lions Eye Bank, Coralville, Iowa
| | - James M Huffman
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Benjamin T Aldrich
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, Iowa; Iowa Lions Eye Bank, Coralville, Iowa
| | - Gregory A Schmidt
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, Iowa; Iowa Lions Eye Bank, Coralville, Iowa
| | - Leandro B C Teixeira
- Department of Pathobiological Sciences, University of Wisconsin-Madison School of Veterinary Medicine, Madison, Wisconsin
| | - Markus H Kuehn
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, Iowa; Center for the Prevention and Treatment of Visual Loss, Iowa City Veterans Affairs Health Care System, Iowa City, Iowa
| | - Mark Yorek
- Center for the Prevention and Treatment of Visual Loss, Iowa City Veterans Affairs Health Care System, Iowa City, Iowa
| | - Mark A Greiner
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, Iowa; Iowa Lions Eye Bank, Coralville, Iowa.
| |
Collapse
|
7
|
Zheng W, Ziemssen F, Suesskind D, Voykov B, Schnichels S. TRPP2 is located in the primary cilia of human non-pigmented ciliary epithelial cells. Graefes Arch Clin Exp Ophthalmol 2024; 262:93-102. [PMID: 37378878 PMCID: PMC10806040 DOI: 10.1007/s00417-023-06150-w] [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: 01/15/2023] [Revised: 05/30/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
PURPOSE Mechanosensitive channels (MSCs) and primary cilium possess a possible relevance for the sensation of intraocular pressure (IOP). However, there is only limited data on their expression and localization in the ciliary body epithelium (CBE). The purpose of this study was to characterize the expression and localization of TRPP2 in a human non-pigmented ciliary epithelial cell (HNPCE) line. METHODS The expression of the TRPP2 was studied by quantitative (q)RT-PCR and in situ hybridization in rat and human tissue. Protein expression and distribution were studied by western blot analysis, immunohistochemistry, and immunoelectron microscopy. Cellular location of TRPP2 was determined in rat and human CBE by immunofluorescence and immunoblot analysis. Electron microscopy studies were conducted to evaluate where and with substructure TRPP2 is localized in the HNPCE cell line. RESULTS The expression of TRPP2 in rat and human non-pigmented ciliary epithelium was detected. TRPP2 was mainly located in nuclei, but also showed a punctate distribution pattern in the cytoplasm of HNPCE of the tissue and the cell line. In HNPCE cell culture, primary cilia did exhibit different length following serum starvation and hydrostatic pressure. TRPP2 was found to be colocalized with these cilia in HNPCE cells. CONCLUSION The expression of TRPP2 and the primary cilium in the CB may indicate a possible role, such as the sensing of hydrostatic pressure, for the regulation of IOP. Functional studies via patch clamp or pharmacological intervention have yet to clarify the relevance for the physiological situation or aqueous humor regulation.
Collapse
Affiliation(s)
- Wenxu Zheng
- Centre for Ophthalmology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Focke Ziemssen
- Centre for Ophthalmology, Eberhard Karls University Tübingen, Tübingen, Germany.
- University Eye Hospital Leipzig, Leipzig, Germany.
- Klinik und Poliklinik für Augenheilkunde, Liebigstr. 10-14, 72072, Leipzig, Germany.
| | - Daniela Suesskind
- Centre for Ophthalmology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Bogomil Voykov
- Centre for Ophthalmology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Sven Schnichels
- Centre for Ophthalmology, Eberhard Karls University Tübingen, Tübingen, Germany
| |
Collapse
|
8
|
Sang Q, Xin C, Yang D, Mu D, Wang N. Effect of Different Postures on Intraocular Pressure in Open-Angle Glaucoma. Ophthalmol Ther 2024; 13:149-160. [PMID: 37924482 PMCID: PMC10776530 DOI: 10.1007/s40123-023-00845-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 10/23/2023] [Indexed: 11/06/2023] Open
Abstract
INTRODUCTION This study aims to investigate the pattern of intraocular pressure (IOP) changes in different postures among patients with open-angle glaucoma (OAG). METHODS A observational study was conducted on a total of 74 patients with OAG (148 eyes). IOP measurements were taken in a variety of positions, including supine, left lateral decubitus, right lateral decubitus, head tilted downwards position with immediate head-up (transient head tilted downwards), seated, seated with head tilted downwards, standing, and walking. Each position was held for 5 min before measurement. In all positions, the patient maintains both eyes looking forward and remains alert. In the head tilted downwards position, the angle of head tilt with respect to the sagittal plane was 30°. RESULTS The overall trend of IOP changes showed a significant decrease with an increase in the position height (r = 0.037, p < 0.001). The IOP was significantly higher in the supine, left lateral decubitus, right lateral decubitus, and head tilted downwards positions than in the seated position (p < 0.001). Compared with the seated position with eyes at primary gaze, IOP decreased significantly when standing (p = 0.008) or walking (p < 0.001). The IOP in the left lateral decubitus and right lateral decubitus was significantly higher than in the supine position (p = 0.008, p = 0.001, respectively). The IOP decreased significantly during walking compared with standing (p < 0.001). CONCLUSIONS The magnitude of IOP strongly correlates with the body position during IOP measurement. The head tilted downwards, supine, left lateral decubitus, and right lateral decubitus positions result in a higher IOP than IOP at the seated position. Patients with OAG can potentially reduce IOP fluctuations by adjusting their daily postures.
Collapse
Affiliation(s)
- Qing Sang
- Department of Ophthalmology, Beijing Tongren Hospital Affiliated to Capital Medical University, Beijing, China
| | - Chen Xin
- Department of Ophthalmology, Beijing Tongren Hospital Affiliated to Capital Medical University, Beijing, China
| | - Diya Yang
- Department of Ophthalmology, Beijing Tongren Hospital Affiliated to Capital Medical University, Beijing, China
| | - Dapeng Mu
- Department of Ophthalmology, Beijing Tongren Hospital Affiliated to Capital Medical University, Beijing, China
| | - Ningli Wang
- Department of Ophthalmology, Beijing Tongren Hospital Affiliated to Capital Medical University, Beijing, China.
- Beijing Ophthalmology Institute, Beijing Tongren Hospital, Capital Medical University, 1 Dongjiaominxiang Street, Dongcheng District, Beijing, 100730, China.
| |
Collapse
|
9
|
Barkander A, Economou MA, Jóhannesson G. Outcomes of iStent Inject Versus Kahook Dual Blade Surgery in Glaucoma Patients Undergoing Cataract Surgery. J Glaucoma 2023; 32:e121-e128. [PMID: 37327470 PMCID: PMC10538610 DOI: 10.1097/ijg.0000000000002243] [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: 12/29/2022] [Accepted: 05/13/2023] [Indexed: 06/18/2023]
Abstract
PRCIS iStent Inject implantation (iStent) or Kahook Dual Blade goniotomy (KDB) in combination with phacoemulsification have a similar IOP-lowering effect in all stages of glaucoma, and medications are significantly reduced, especially after KDB. PURPOSE To compare the 2-year efficacy and safety of iStent or KDB in combination with phacoemulsification in eyes with mild to advanced open angle glaucoma. METHODS A retrospective chart review of 153 patients that received iStent or KDB in combination with phacoemulsification at a single center between March 2019 and August 2020. The main outcome parameters at 2 years were: (1) intraocular pressure (IOP)-reduction ≥20%, with a postoperative IOP ≤18 mm Hg, and (2) a reduction of ≥1 medication. Results were stratified by glaucoma grade. RESULTS After 2 years, mean IOP was reduced from 20.3±6.1 to 14.2±4.1 mm Hg in the phaco-iStent group ( P <0.001) and from 20.1±6.1 to 14.7±3.6 mm Hg in the phaco-KDB group ( P <0.001). The mean number of medications was reduced from 3.0±0.9 to 2.6±1.1 in the Phaco-iStent group ( P =0.001) and from 2.3±1.0 to 1.5±1.3 in the Phaco-KDB group ( P <0.001). Success regarding IOP-reduction ≥20% with a postoperative IOP ≤18 mm Hg was met by 46% in the phaco-iStent group and by 51% in the phaco-KDB group. A reduction of ≥1 medication was met by 32% in the phaco-iStent group and by 53% in the phaco-KDB group ( P =0.013). Eyes with mild to moderate and advanced glaucoma responded equally well to the success criteria. CONCLUSIONS iStent and KDB, in combination with phacoemulsification, both lowered IOP effectively in all stages of glaucoma. More medications were reduced after KDB, suggesting that it may be a more effective procedure compared with iStent.
Collapse
Affiliation(s)
- Anna Barkander
- Department of Clinical Sciences, Ophthalmology, Umeå University, Umeå, Sweden
| | - M A Economou
- Division of Ophthalmology and Vision, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Ophthalmology, Capio Sophiahemmet Hospital, Stockholm, Sweden
| | - Gauti Jóhannesson
- Department of Clinical Sciences, Ophthalmology, Umeå University, Umeå, Sweden
- Wallenberg Centre for Molecular Medicine, Umeå University, Umeå, Sweden
- Department of Ophthalmology, University of Iceland, Reykjavik, Iceland
| |
Collapse
|
10
|
Sikiric P, Kokot A, Kralj T, Zlatar M, Masnec S, Lazic R, Loncaric K, Oroz K, Sablic M, Boljesic M, Antunovic M, Sikiric S, Strbe S, Stambolija V, Beketic Oreskovic L, Kavelj I, Novosel L, Zubcic S, Krezic I, Skrtic A, Jurjevic I, Boban Blagaic A, Seiwerth S, Staresinic M. Stable Gastric Pentadecapeptide BPC 157-Possible Novel Therapy of Glaucoma and Other Ocular Conditions. Pharmaceuticals (Basel) 2023; 16:1052. [PMID: 37513963 PMCID: PMC10385428 DOI: 10.3390/ph16071052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 07/01/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023] Open
Abstract
Recently, stable gastric pentadecapeptide BPC 157 therapy by activation of collateral pathways counteracted various occlusion/occlusion-like syndromes, vascular, and multiorgan failure, and blood pressure disturbances in rats with permanent major vessel occlusion and similar procedures disabling endothelium function. Thereby, we revealed BPC 157 cytoprotective therapy with strong vascular rescuing capabilities in glaucoma therapy. With these capabilities, BPC 157 therapy can recover glaucomatous rats, normalize intraocular pressure, maintain retinal integrity, recover pupil function, recover retinal ischemia, and corneal injuries (i.e., maintained transparency after complete corneal abrasion, corneal ulceration, and counteracted dry eye after lacrimal gland removal or corneal insensitivity). The most important point is that in glaucomatous rats (three of four episcleral veins cauterized) with high intraocular pressure, all BPC 157 regimens immediately normalized intraocular pressure. BPC 157-treated rats exhibited normal pupil diameter, microscopically well-preserved ganglion cells and optic nerve presentation, normal fundus presentation, nor- mal retinal and choroidal blood vessel presentation, and normal optic nerve presentation. The one episcleral vein rapidly upgraded to accomplish all functions in glaucomatous rats may correspond with occlusion/occlusion-like syndromes of the activated rescuing collateral pathway (azygos vein direct blood flow delivery). Normalized intraocular pressure in glaucomatous rats corresponded to the counteracted intra-cranial (superior sagittal sinus), portal, and caval hypertension, and aortal hypotension in occlusion/occlusion-like syndromes, were all attenuated/eliminated by BPC 157 therapy. Furthermore, given in other eye disturbances (i.e., retinal ischemia), BPC 157 instantly breaks a noxious chain of events, both at an early stage and an already advanced stage. Thus, we further advocate BPC 157 as a therapeutic agent in ocular disease.
Collapse
Affiliation(s)
- Predrag Sikiric
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Antonio Kokot
- Department of Anatomy and Neuroscience, Faculty of Medicine, J.J. Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Tamara Kralj
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Mirna Zlatar
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Sanja Masnec
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Ratimir Lazic
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Kristina Loncaric
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Katarina Oroz
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Marko Sablic
- Department of Anatomy and Neuroscience, Faculty of Medicine, J.J. Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Marta Boljesic
- Department of Anatomy and Neuroscience, Faculty of Medicine, J.J. Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Marko Antunovic
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Suncana Sikiric
- Department of Pathology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Sanja Strbe
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Vasilije Stambolija
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | | | - Ivana Kavelj
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Luka Novosel
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Slavica Zubcic
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Ivan Krezic
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Anita Skrtic
- Department of Pathology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Ivana Jurjevic
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Alenka Boban Blagaic
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Sven Seiwerth
- Department of Pathology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Mario Staresinic
- Department of Surgery, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| |
Collapse
|
11
|
Sharif NA. Recently Approved Drugs for Lowering and Controlling Intraocular Pressure to Reduce Vision Loss in Ocular Hypertensive and Glaucoma Patients. Pharmaceuticals (Basel) 2023; 16:791. [PMID: 37375739 DOI: 10.3390/ph16060791] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 05/17/2023] [Accepted: 05/19/2023] [Indexed: 06/29/2023] Open
Abstract
Serious vision loss occurs in patients affected by chronically raised intraocular pressure (IOP), a characteristic of many forms of glaucoma where damage to the optic nerve components causes progressive degeneration of retinal and brain neurons involved in visual perception. While many risk factors abound and have been validated for this glaucomatous optic neuropathy (GON), the major one is ocular hypertension (OHT), which results from the accumulation of excess aqueous humor (AQH) fluid in the anterior chamber of the eye. Millions around the world suffer from this asymptomatic and progressive degenerative eye disease. Since clinical evidence has revealed a strong correlation between the reduction in elevated IOP/OHT and GON progression, many drugs, devices, and surgical techniques have been developed to lower and control IOP. The constant quest for new pharmaceuticals and other modalities with superior therapeutic indices has recently yielded health authority-approved novel drugs with unique pharmacological signatures and mechanism(s) of action and AQH drainage microdevices for effectively and durably treating OHT. A unique nitric oxide-donating conjugate of latanoprost, an FP-receptor prostaglandin (PG; latanoprostene bunod), new rho kinase inhibitors (ripasudil; netarsudil), a novel non-PG EP2-receptor-selective agonist (omidenepag isopropyl), and a form of FP-receptor PG in a slow-release intracameral implant (Durysta) represent the additions to the pharmaceutical toolchest to mitigate the ravages of OHT. Despite these advances, early diagnosis of OHT and glaucoma still lags behind and would benefit from further concerted effort and attention.
Collapse
Affiliation(s)
- Najam A Sharif
- Eye-APC Duke-NUS Medical School, Singapore 169856, Singapore
- Singapore Eye Research Institute, Singapore 169856, Singapore
- Department of Pharmacology and Neuroscience, University of North Texas Health Sciences Center, Fort Worth, TX 76107, USA
- Department of Pharmacy Sciences, Creighton University, Omaha, NE 68178, USA
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Texas Southern University, Houston, TX 77004, USA
- Imperial College of Science and Technology, St. Mary's Campus, London SW7 2BX, UK
- Institute of Ophthalmology, University College London, London WC1E 6BT, UK
| |
Collapse
|
12
|
Gao XR, Chiariglione M, Choquet H, Arch AJ. 10 Years of GWAS in intraocular pressure. Front Genet 2023; 14:1130106. [PMID: 37124618 PMCID: PMC10130654 DOI: 10.3389/fgene.2023.1130106] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 04/05/2023] [Indexed: 05/02/2023] Open
Abstract
Intraocular pressure (IOP) is the only modifiable risk factor for glaucoma, the leading cause of irreversible blindness worldwide. In this review, we summarize the findings of genome-wide association studies (GWASs) of IOP published in the past 10 years and prior to December 2022. Over 190 genetic loci and candidate genes associated with IOP have been uncovered through GWASs, although most of these studies were conducted in subjects of European and Asian ancestries. We also discuss how these common variants have been used to derive polygenic risk scores for predicting IOP and glaucoma, and to infer causal relationship with other traits and conditions through Mendelian randomization. Additionally, we summarize the findings from a recent large-scale exome-wide association study (ExWAS) that identified rare variants associated with IOP in 40 novel genes, six of which are drug targets for clinical treatment or are being evaluated in clinical trials. Finally, we discuss the need for future genetic studies of IOP to include individuals from understudied populations, including Latinos and Africans, in order to fully characterize the genetic architecture of IOP.
Collapse
Affiliation(s)
- Xiaoyi Raymond Gao
- Department of Ophthalmology and Visual Sciences, The Ohio State University, Columbus, OH, United States
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, United States
- Division of Human Genetics, The Ohio State University, Columbus, OH, United States
| | - Marion Chiariglione
- Department of Ophthalmology and Visual Sciences, The Ohio State University, Columbus, OH, United States
| | - Hélène Choquet
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, United States
| | - Alexander J. Arch
- Department of Ophthalmology and Visual Sciences, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
13
|
Sharif NA, Odani-Kawabata N, Lu F, Pinchuk L. FP and EP2 prostanoid receptor agonist drugs and aqueous humor outflow devices for treating ocular hypertension and glaucoma. Exp Eye Res 2023; 229:109415. [PMID: 36803996 DOI: 10.1016/j.exer.2023.109415] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/21/2022] [Accepted: 02/08/2023] [Indexed: 02/21/2023]
Abstract
Prostaglandin (PG) receptors represent important druggable targets due to the many diverse actions of PGs in the body. From an ocular perspective, the discovery, development, and health agency approvals of prostaglandin F (FP) receptor agonists (FPAs) have revolutionized the medical treatment of ocular hypertension (OHT) and glaucoma. FPAs, such as latanoprost, travoprost, bimatoprost, and tafluprost, powerfully lower and control intraocular pressure (IOP), and became first-line therapeutics to treat this leading cause of blindness in the late 1990s to early 2000s. More recently, a latanoprost-nitric oxide (NO) donor conjugate, latanoprostene bunod, and a novel FP/EP3 receptor dual agonist, sepetaprost (ONO-9054 or DE-126), have also demonstrated robust IOP-reducing activity. Moreover, a selective non-PG prostanoid EP2 receptor agonist, omidenepag isopropyl (OMDI), was discovered, characterized, and has been approved in the United States, Japan and several other Asian countries for treating OHT/glaucoma. FPAs primarily enhance uveoscleral (UVSC) outflow of aqueous humor (AQH) to reduce IOP, but cause darkening of the iris and periorbital skin, uneven thickening and elongation of eyelashes, and deepening of the upper eyelid sulcus during chronic treatment. In contrast, OMDI lowers and controls IOP by activation of both the UVSC and trabecular meshwork outflow pathways, and it has a lower propensity to induce the aforementioned FPA-induced ocular side effects. Another means to address OHT is to physically promote the drainage of the AQH from the anterior chamber of the eye of patients with OHT/glaucoma. This has successfully been achieved by the recent approval and introduction of miniature devices into the anterior chamber by minimally invasive glaucoma surgeries. This review covers the three major aspects mentioned above to highlight the etiology of OHT/glaucoma, and the pharmacotherapeutics and devices that can be used to combat this blinding ocular disease.
Collapse
Affiliation(s)
- Najam A Sharif
- Ophthalmology Innovation Center, Santen Inc., Emeryville, CA, USA; Singapore Eye Research Institute, Singapore; Eye-ACP Duke-National University of Singapore Medical School, Singapore; Department of Pharmacology and Neuroscience, University of North Texas Health Sciences Center, Fort Worth, TX, USA; Department of Pharmacy Sciences, Creighton University, Omaha, NE, USA; Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Texas Southern University, Houston, TX, USA; Imperial College of Science and Technology, St. Mary's Campus, London, UK; Institute of Ophthalmology, University College London, London, UK.
| | | | - Fenghe Lu
- Product Development Division, Santen Inc., Emeryville, CA, USA
| | - Leonard Pinchuk
- Ophthalmology Innovation Center, Santen Inc., Emeryville, CA, USA; Biomedical Engineering Department, University of Miami, Miami, FL, USA
| |
Collapse
|
14
|
Mechanical Stretch Activates TRPV4 and Hemichannel Responses in the Nonpigmented Ciliary Epithelium. Int J Mol Sci 2023; 24:ijms24021673. [PMID: 36675184 PMCID: PMC9865367 DOI: 10.3390/ijms24021673] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/05/2023] [Accepted: 01/12/2023] [Indexed: 01/19/2023] Open
Abstract
Previously, we reported a mechanosensitive ion channel, TRPV4, along with functional connexin hemichannels on the basolateral surface of the ocular nonpigmented ciliary epithelium (NPE). In the lens, TRPV4-mediated hemichannel opening is part of a feedback loop that senses and respond to swelling. The present study was undertaken to test the hypothesis that TRPV4 and hemichannels in the NPE respond to a mechanical stimulus. Porcine NPE cells were cultured on flexible membranes to study effects of cyclic stretch and ATP release was determined by a luciferase assay. The uptake of propidium iodide (PI) was measured as an indicator of hemichannel opening. NPE cells subjected to cyclic stretch for 1-10 min (10%, 0.5 Hz) displayed a significant increase in ATP release into the bathing medium. In studies where PI was added to the bathing medium, the same stretch stimulus increased cell PI uptake. The ATP release and PI uptake responses to stretch both were prevented by a TRPV4 antagonist, HC067047 (10 µM), and a connexin mimetic peptide, Gap 27 (200µm). In the absence of a stretch stimulus, qualitatively similar ATP release and PI uptake responses were observed in cells exposed to the TRPV4 agonist GSK1016790A (10 nM), and Gap 27 prevented the responses. Cells subjected to an osmotic swelling stimulus (hypoosmotic medium: 200 mOsm) also displayed a significant increase in ATP release and PI uptake and the responses were abolished by TRPV4 inhibition. The findings point to TRPV4-dependent connexin hemichannel opening in response to mechanical stimulus. The TRPV4-hemichannel mechanism may act as a mechanosensor that facilitates the release of ATP and possibly other autocrine or paracrine signaling molecules that influence fluid (aqueous humor) secretion by the NPE.
Collapse
|
15
|
Wostyn P, Killer HE. Normal-Tension Glaucoma: A Glymphopathy? Eye Brain 2023; 15:37-44. [PMID: 37056720 PMCID: PMC10086217 DOI: 10.2147/eb.s401306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 03/07/2023] [Indexed: 04/15/2023] Open
Abstract
Glaucoma is one of the main causes of irreversible blindness in the world. The most common form, primary open-angle glaucoma, is an optic neuropathy that is characterized by a progressive loss of retinal ganglion cells and their axons, leading to structural changes in the optic nerve head and associated visual field defects. Elevated intraocular pressure remains the most important modifiable risk factor for primary open-angle glaucoma. However, a significant proportion of patients develop glaucomatous damage in the absence of increased intraocular pressure, a condition known as normal-tension glaucoma (NTG). The pathophysiology underlying NTG remains unclear. Several studies have revealed that vascular and cerebrospinal fluid (CSF) factors may play significant roles in the development of NTG. Vascular failure caused by functional or structural abnormalities, and compartmentation of the optic nerve subarachnoid space with disturbed CSF dynamics have been shown to be associated with NTG. In the present article, based on the concept of the glymphatic system and observations in patients with NTG, we hypothesize that failure of fluid transport via the glymphatic pathway in the optic nerve may be involved in the pathogenesis of some if not many cases of NTG. According to this hypothesis, vascular and CSF factors may share reduced glymphatic transport and perivascular waste clearance in the optic nerve as a final common pathway leading to the development of NTG. In addition, we speculate that some cases of NTG may reflect glymphatic dysfunction in natural brain aging and central nervous system diseases, such as Alzheimer's disease. Clearly, further studies are needed to gain additional insight into the relative contribution of these factors and conditions to reduced glymphatic transport in the optic nerve.
Collapse
Affiliation(s)
- Peter Wostyn
- Department of Psychiatry, PC Sint-Amandus, Beernem, Belgium
- Correspondence: Peter Wostyn, Department of Psychiatry, PC Sint-Amandus, Reigerlostraat 10, Beernem, 8730, Belgium, Tel +32-472713719, Fax +32-50-819720, Email
| | - Hanspeter Esriel Killer
- Department of Biomedicine, University of Basel, Basel, Switzerland
- Augenärzte Zentrum Aarau, Aarau, Switzerland
| |
Collapse
|
16
|
Barkander A, Economou MA, Jóhannesson G. Kahook Dual-Blade Goniotomy with and without Phacoemulsification in Medically Uncontrolled Glaucoma. Clin Ophthalmol 2023; 17:1385-1394. [PMID: 37204995 PMCID: PMC10187655 DOI: 10.2147/opth.s409375] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 04/20/2023] [Indexed: 05/21/2023] Open
Abstract
Purpose To evaluate the 2-year efficacy and safety of Kahook dual-blade (KDB) goniotomy in patients with medically uncontrolled glaucoma. Methods This was a retrospective case-series study of 90 consecutive patients with primary open-angle glaucoma (POAG) or pseudoexfoliation glaucoma (PEXG) that underwent KDB goniotomy alone (KDB-alone group) or KDB goniotomy in combination with phacoemulsification (KDB-phaco group) during 2019-2020. All patients were uncontrolled on three or more medications. Surgical success was defined as an IOP reduction ≥20% and/or a reduction of one or more medications at 24 months. We also report IOP levels and number of medications from baseline to 24 months, as well as the need for further glaucoma interventions. Results At 24 months, mean IOP had reduced from 24.8±8.3 to 15.0±5.3 mmHg in the KDB-alone group (P<0.001) and from 22.3±5.8 to 13.9±3.0 mmHg in the KDB-phaco group (P<0.001). Medications had reduced from 3.5±0.6 to 3.1±0.9 in the KDB-alone group (P=0.047) and from 3.3±0.5 to 2.3±1.1 in the KDB-phaco group (P<0.001). An IOP reduction ≥20% and/or a reduction with one or more medications was achieved by 47% of eyes in the KDB-alone group and by 76% of eyes in the KDB-phaco group. Eyes with PEXG and POAG responded equally well to the success criteria. During the 24-month follow-up, additional glaucoma surgery or transscleral photocoagulation was performed in 28% of eyes in the KDB-alone group and in 12% of eyes in the KDB-phaco group. Conclusion In patients with medically uncontrolled glaucoma, KDB had a significant IOP-lowering effect after 24 months, but success rates were higher when KDB was performed in combination with cataract surgery compared to stand-alone treatment.
Collapse
Affiliation(s)
- Anna Barkander
- Department of Clinical Sciences, Ophthalmology, Umeå University, Umeå, Sweden
- Correspondence: Anna Barkander, Department of Clinical Sciences, Ophthalmology, Umeå University, Umeå, SE-901 85, Sweden, Tel +46 63 15 30 00, Email
| | - Mario A Economou
- Division of Ophthalmology and Vision, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Ophthalmology, Capio Sophiahemmet Hospital, Stockholm, Sweden
| | - Gauti Jóhannesson
- Department of Clinical Sciences, Ophthalmology, Umeå University, Umeå, Sweden
- Wallenberg Centre for Molecular Medicine, Umeå University, Umeå, Sweden
- Department of Ophthalmology, University of Iceland, Reykjavik, Iceland
| |
Collapse
|
17
|
Ouyang J, Sun W, Shen H, Liu X, Wu Y, Jiang H, Li X, Wang Y, Jiang Y, Li S, Xiao X, Hejtmancik JF, Tan Z, Zhang Q. Truncation mutations in MYRF underlie primary angle closure glaucoma. Hum Genet 2023; 142:103-123. [PMID: 36129575 DOI: 10.1007/s00439-022-02487-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 09/05/2022] [Indexed: 01/18/2023]
Abstract
Mutations in myelin regulatory factor (MYRF), a gene mapped to 11q12-q13.3, are responsible for autosomal dominant high hyperopia and seem to be associated with angle closure glaucoma, which is one of the leading causes of irreversible blindness worldwide. Whether there is a causal link from the MYRF mutations to the pathogenesis of primary angle-closure glaucoma (PACG) remains unclear at this time. Six truncation mutations, including five novel and one previously reported, in MYRF are identified in seven new probands with hyperopia, of whom all six adults have glaucoma, further confirming the association of MYRF mutations with PACG. Immunofluorescence microscopy demonstrates enriched expression of MYRF in the ciliary body and ganglion cell layer in humans and mice. Myrfmut/+ mice have elevated IOP and fewer ganglion cells along with thinner retinal nerve fiber layer with ganglion cell layer than wild-type. Transcriptome sequencing of Myrfmut/+ retinas shows downregulation of Dnmt3a, a gene previously associated with PACG. Co-immunoprecipitation demonstrates a physical association of DNMT3A with MYRF. DNA methylation sequencing identifies several glaucoma-related cell events in Myrfmut/+ retinas. The interaction between MYRF and DNMT3A underlies MYRF-associated PACG and provides clues for pursuing further investigation into the pathogenesis of PACG and therapeutic target.
Collapse
Affiliation(s)
- Jiamin Ouyang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, 54 Xianlie Road, Guangzhou, 510060, China
| | - Wenmin Sun
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, 54 Xianlie Road, Guangzhou, 510060, China
| | - Huangxuan Shen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, 54 Xianlie Road, Guangzhou, 510060, China
| | - Xing Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, 54 Xianlie Road, Guangzhou, 510060, China
| | - Yingchen Wu
- Department of Gynecology and Obstetrics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hongmei Jiang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, 54 Xianlie Road, Guangzhou, 510060, China
| | - Xueqing Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, 54 Xianlie Road, Guangzhou, 510060, China
| | - Yingwei Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, 54 Xianlie Road, Guangzhou, 510060, China
| | - Yi Jiang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, 54 Xianlie Road, Guangzhou, 510060, China
| | - Shiqiang Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, 54 Xianlie Road, Guangzhou, 510060, China
| | - Xueshan Xiao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, 54 Xianlie Road, Guangzhou, 510060, China
| | - J Fielding Hejtmancik
- Molecular Ophthalmic Genetics Section, Ophthalmic Genetics and Visual Function Branch, National Eye Institute, Rockville, MD, 20852, USA.
| | - Zhiqun Tan
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA.
| | - Qingjiong Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, 54 Xianlie Road, Guangzhou, 510060, China.
| |
Collapse
|
18
|
Asaoka R, Obana A, Murata H, Fujino Y, Omoto T, Aoki S, Muto S, Takayanagi Y, Inoue T, Tanito M. The Association Between Age and Systemic Variables and the Longitudinal Trend of Intraocular Pressure in a Large-Scale Health Examination Cohort. Invest Ophthalmol Vis Sci 2022; 63:22. [PMID: 36301531 PMCID: PMC9624273 DOI: 10.1167/iovs.63.11.22] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose The detailed effects of age and systemic factors on intraocular pressure (IOP) have not been fully understood because of the lack of a large-scale longitudinal investigation. This study aimed to investigate the effect of various systemic factors on the longitudinal change of IOP. Methods There were a total of 20,909 eyes of 10,471 subjects from a health checkup cohort that were followed up for systemic factors: (i) age at baseline, (ii) sex, (iii) time series body mass index (BMI), (iv) time series smoking habits, (v) time series systolic and diastolic blood pressures (SBP and DBP), and (vi) time series 19 blood examinations (all of the time series data was acquired at each annual visit), along with IOP annually for at least 8 years. Then the longitudinal effect of the systemic factors on the change of IOP was investigated. Results IOP significantly decreased by −0.084 mm Hg/year. BMI, SBP, DBP, smoking habits, total triglyceride, high-density lipoprotein cholesterol, low-density lipoprotein cholesterol, and glycosylated hemoglobin A1c were not significantly associated with the change of IOP. Higher values of age, aspartate aminotransferase, hemoglobin, platelet, and calcium were suggested to be significantly associated with the decrease of IOP, whereas higher alanine aminotransferase, guanosine triphosphate, white blood cell count, red blood cell count, and female gender were significantly associated with the increase of IOP. Conclusions Age, aspartate aminotransferase, hemoglobin, platelet, calcium, alanine aminotransferase, guanosine triphosphate, white blood cell count, red blood cell count, and gender were the systemic variables significantly associated with the change of IOP.
Collapse
Affiliation(s)
- Ryo Asaoka
- Department of Ophthalmology, Seirei Hamamatsu General Hospital, Shizuoka, Hamamatsu, Japan
- Seirei Christopher University, Shizuoka, Hamamatsu, Japan
- Department of Ophthalmology, The University of Tokyo, Tokyo, Japan
- Nanovision Research Division, Research Institute of Electronics, Shizuoka University, Shizuoka Japan
- The Graduate School for the Creation of New Photonics Industries, Shizuoka Japan
| | - Akira Obana
- Department of Ophthalmology, Seirei Hamamatsu General Hospital, Shizuoka, Hamamatsu, Japan
- Hamamatsu BioPhotonics Innovation Chair, Institute for Medical Photonics Research, Preeminent Medical Photonics Education & Research Center, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Hiroshi Murata
- Department of Ophthalmology, The University of Tokyo, Tokyo, Japan
- Department of Ophthalmology, National Center for Global Health and Medicine, Shinjuku, Tokyo, Japan
| | - Yuri Fujino
- Department of Ophthalmology, Seirei Hamamatsu General Hospital, Shizuoka, Hamamatsu, Japan
- Department of Ophthalmology, Shimane University Faculty of Medicine, Izumo, Japan
| | - Takashi Omoto
- Department of Ophthalmology, The University of Tokyo, Tokyo, Japan
| | - Shuichiro Aoki
- Department of Ophthalmology, Sapporo City General Hospital, Sapporo, Japan
| | - Shigetaka Muto
- Seirei Center for Health Promotion and Preventive Medicine, Shizuoka, Hamamatsu, Japan
| | - Yuji Takayanagi
- Department of Ophthalmology, Seirei Hamamatsu General Hospital, Shizuoka, Hamamatsu, Japan
- Department of Ophthalmology, Shimane University Faculty of Medicine, Izumo, Japan
| | - Tatsuya Inoue
- Department of Ophthalmology and Micro-Technology, Yokohama City University, Kanagawa, Japan
| | - Masaki Tanito
- Department of Ophthalmology, Shimane University Faculty of Medicine, Izumo, Japan
| |
Collapse
|
19
|
Hann CR, Bentley MD, Vercnocke A, Roy Chowdhury U, Fautsch MP. Evaluation of neural innervation in the human conventional outflow pathway distal to Schlemm's canal. Exp Eye Res 2022; 221:109132. [PMID: 35636488 PMCID: PMC10493174 DOI: 10.1016/j.exer.2022.109132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 04/29/2022] [Accepted: 05/23/2022] [Indexed: 11/26/2022]
Abstract
The distal outflow pathway of the human eye consists of the outer wall of Schlemm's canal, collector channels, and the deep-scleral, mid-scleral and episcleral vessels. It is the last region of transit for aqueous humor before returning to the venous system. While the trabecular meshwork, scleral spur, and inner wall of Schlemm's canal have been extensively analyzed to define their contributions to aqueous outflow, the role of the distal outflow pathway is not completely understood. Collector channels, emanating from Schlemm's canal were previously thought to be passive conduits for aqueous humor. However, recent studies have shown many collector channels contain flap-like appendages which move with changes in pressure. These findings, along with studies demonstrating innervation of episcleral vessels, have led to questions regarding whether other structures in the distal outflow pathway are under neural regulation and how this may influence aqueous humor outflow. This study evaluates the innervation of the outer wall of Schlemm's canal and collector channels, along with the deep-scleral, mid-scleral and episcleral vasculature with microcomputed tomography and 3-dimensional reconstruction, correlative light microscopy, immunohistochemistry, and transmission electron microscopy. Peripheral, autonomic, and sensory nerve fibers were found to be present adjacent to Schlemm's canal outer wall endothelium, collector channel endothelium, and in the different regions of the distal outflow vasculature. Nerves were more commonly identified in regions that contained collector channels when compared to regions without collector channels. These findings regarding the neural anatomy suggest an active neural regulation of aqueous humor outflow throughout the proximal and distal regions of the conventional outflow pathway.
Collapse
Affiliation(s)
- Cheryl R Hann
- Department of Ophthalmology, Mayo Clinic, Rochester, MN, USA.
| | | | - Andrew Vercnocke
- Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA.
| | | | | |
Collapse
|
20
|
Detailed 3D micro-modeling of rat aqueous drainage channels based on two-photon imaging: simulating aqueous humor through trabecular meshwork and Schlemm’s canal by two-way fluid structure interaction approach. Med Biol Eng Comput 2022; 60:1915-1927. [DOI: 10.1007/s11517-022-02580-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 04/22/2022] [Indexed: 10/18/2022]
|
21
|
Sharif NA. Degeneration of retina-brain components and connections in glaucoma: Disease causation and treatment options for eyesight preservation. CURRENT RESEARCH IN NEUROBIOLOGY 2022; 3:100037. [PMID: 36685768 PMCID: PMC9846481 DOI: 10.1016/j.crneur.2022.100037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 03/30/2022] [Accepted: 04/01/2022] [Indexed: 01/25/2023] Open
Abstract
Eyesight is the most important of our sensory systems for optimal daily activities and overall survival. Patients who experience visual impairment due to elevated intraocular pressure (IOP) are often those afflicted with primary open-angle glaucoma (POAG) which slowly robs them of their vision unless treatment is administered soon after diagnosis. The hallmark features of POAG and other forms of glaucoma are damaged optic nerve, retinal ganglion cell (RGC) loss and atrophied RGC axons connecting to various brain regions associated with receipt of visual input from the eyes and eventual decoding and perception of images in the visual cortex. Even though increased IOP is the major risk factor for POAG, the disease is caused by many injurious chemicals and events that progress slowly within all components of the eye-brain visual axis. Lowering of IOP mitigates the damage to some extent with existing drugs, surgical and device implantation therapeutic interventions. However, since multifactorial degenerative processes occur during aging and with glaucomatous optic neuropathy, different forms of neuroprotective, nutraceutical and electroceutical regenerative and revitalizing agents and processes are being considered to combat these eye-brain disorders. These aspects form the basis of this short review article.
Collapse
Affiliation(s)
- Najam A. Sharif
- Duke-National University of Singapore Medical School, Singapore,Singapore Eye Research Institute (SERI), Singapore,Department of Pharmacology and Neuroscience, University of North Texas Health Sciences Center, Fort Worth, Texas, USA,Department of Pharmaceutical Sciences, Texas Southern University, Houston, TX, USA,Department of Surgery & Cancer, Imperial College of Science and Technology, St. Mary's Campus, London, UK,Department of Pharmacy Sciences, School of School of Pharmacy and Health Professions, Creighton University, Omaha, NE, USA,Ophthalmology Innovation Center, Santen Incorporated, 6401 Hollis Street (Suite #125), Emeryville, CA, 94608, USA,Ophthalmology Innovation Center, Santen Incorporated, 6401 Hollis Street (Suite #125), Emeryville, CA, 94608, USA.
| |
Collapse
|
22
|
Almarzouki N, Bafail SA, Danish DH, Algethami SR, Shikdar N, Ashram S, Roblah T. The Impact of Systemic Health Parameters on Intraocular Pressure in the Western Region of Saudi Arabia. Cureus 2022; 14:e25217. [PMID: 35747032 PMCID: PMC9213110 DOI: 10.7759/cureus.25217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/22/2022] [Indexed: 11/21/2022] Open
Abstract
Background A normal intraocular pressure (IOP) is essential for maintaining the normal structure and function of the eyes. Furthermore, an elevated IOP is a known risk factor for glaucoma. As the results from studies addressing the relationship between IOP and systemic health parameters are conflicting, researchers have not reached a consensus. This study aimed to evaluate the relationship between IOP and health parameters among non-glaucomatous participants in the western region of Saudi Arabia. Method We retrospectively reviewed the medical records of 255 participants aged 20 years and above who had not received any medical treatment for ocular hypertension or glaucoma at King Abdulaziz University Hospital (KAUH), Jeddah, Saudi Arabia, from January 2019 to April 2021. The patients were categorized into age groups, divided by decades (ranging from 20-29 years to 80+ years); the most prevalent age group was 60-69 years. The data were entered using Microsoft Excel 2016 (Microsoft Corporation, Redmond, Washington), and Statistical Package for the Social Sciences (SPSS) software was used for univariate analysis. The relationship between continuous variables was analyzed by the Pearson correlation coefficient. The differences between continuous and categorical variables were assessed by the t-test and one-way analysis of variance (ANOVA) test, respectively. Results The mean (standard deviation) IOP in the right and the left eyes was 15.7 mmHg (4.0) and 15.6 mmHg (3.9), respectively. There were no significant associations between IOP and lipid profile parameters (p > 0.05). There was a statistically significant difference in the right IOP, in relation to the age groups (p = 0.006). Moreover, the mean IOP in the left eye was significantly higher among patients with diabetes than in the patients without diabetes (p = 0.007) as well as in patients with hypertension than in the patients without hypertension (p = 0.023). Conclusion The effect of total cholesterol, triglyceride (TG), low-density lipoprotein (LDL), high-density lipoprotein (HDL), BMI, and sex on IOP could not be established in our study. However, over the past years, people's diets have incorporated higher cholesterol and fat content, leading to higher BMI levels. Therefore, further studies of the association between BMI and IOP are critical to determine if BMI is certainly a significant risk factor for increased IOP and glaucoma.
Collapse
|
23
|
Kim Y, Yang J, Kim JY, Lee JM, Son WC, Moon BG. HL3501, a Novel Selective A3 Adenosine Receptor Antagonist, Lowers Intraocular Pressure (IOP) in Animal Glaucoma Models. Transl Vis Sci Technol 2022; 11:30. [PMID: 35191964 PMCID: PMC8883152 DOI: 10.1167/tvst.11.2.30] [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: 11/24/2022] Open
Abstract
Purpose The A3 adenosine receptor (A3AR) is a known therapeutic target for glaucoma treatment. In this study, we developed HL3501 and examined its selectivity profile and in vitro and in vivo effects. Methods For the rabbit model, intraocular pressure (IOP) was increased by laser photocoagulation of the trabecular meshwork (TM). The rabbits were then topically treated with HL3501, latanoprost, timolol, or vehicle for 3 weeks. For the mouse model, HL3501, latanoprost, or vehicle was administered following induced IOP elevation by dexamethasone (Dex). The IOP of all rabbits and mice was measured. Electroretinography was performed on both eyes of dark-adapted anesthetized mice on days 0 and 21. The mice's eyes were enucleated at the end of the treatment for immunofluorescence staining. Results HL3501 was highly specific to the A3AR and inhibitory of A3AR function. In the rabbit glaucoma model, HL3501 and latanoprost significantly decreased the IOP. In the Dex-treated mouse model, HL3501 and latanoprost significantly decreased the IOP and increased the b-wave amplitude as compared with the vehicle treatment. HL3501 and latanoprost also inhibited fibronectin and α-smooth muscle actin expression induced by Dex treatment. Conclusions HL3501 had effects similar to those of latanoprost in reducing ocular hypertension in animal models. HL3501 could be used as a novel approach to treat glaucoma. Translational Relevance HL3501 is a novel preclinical compound targeting the A3 adenosine receptor, which may also be a new treatment option to fill the unmet needs of many glaucoma patients.
Collapse
Affiliation(s)
- Yunhee Kim
- Handok Pharmaceuticals, Seoul, Republic of Korea.,Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Jaewook Yang
- T2B Infrastructure Center for Ocular Disease, Busan Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea
| | - Jee Young Kim
- T2B Infrastructure Center for Ocular Disease, Busan Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea
| | - Jang Mi Lee
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Woo Chan Son
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | | |
Collapse
|
24
|
Delamere NA, Shahidullah M. Ion Transport Regulation by TRPV4 and TRPV1 in Lens and Ciliary Epithelium. Front Physiol 2022; 12:834916. [PMID: 35173627 PMCID: PMC8841554 DOI: 10.3389/fphys.2021.834916] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 12/27/2021] [Indexed: 02/02/2023] Open
Abstract
Aside from a monolayer of epithelium at the anterior surface, the lens is formed by tightly compressed multilayers of fiber cells, most of which are highly differentiated and have a limited capacity for ion transport. Only the anterior monolayer of epithelial cells has high Na, K-ATPase activity. Because the cells are extensively coupled, the lens resembles a syncytium and sodium-potassium homeostasis of the entire structure is largely dependent on ion transport by the epithelium. Here we describe recent studies that suggest TRPV4 and TRPV1 ion channels activate signaling pathways that play an important role in matching epithelial ion transport activity with needs of the lens cell mass. A TRPV4 feedback loop senses swelling in the fiber mass and increases Na, K-ATPase activity to compensate. TRPV4 channel activation in the epithelium triggers opening of connexin hemichannels, allowing the release of ATP that stimulates purinergic receptors in the epithelium and results in the activation of Src family tyrosine kinases (SFKs) and SFK-dependent increase of Na, K-ATPase activity. A separate TRPV1 feedback loop senses shrinkage in the fiber mass and increases NKCC1 activity to compensate. TRPV1 activation causes calcium-dependent activation of a signaling cascade in the lens epithelium that involves PI3 kinase, ERK, Akt and WNK. TRPV4 and TRPV1 channels are also evident in the ciliary body where Na, K-ATPase is localized on one side of a bilayer in which two different cell types, non-pigmented and pigmented ciliary epithelium, function in a coordinated manner to secrete aqueous humor. TRPV4 and TRPV1 may have a role in maintenance of cell volume homeostasis as ions and water move through the bilayer.
Collapse
|
25
|
Taiyab A, Akula M, Dham J, Deschamps P, Sheardown H, Williams T, Borrás T, West-Mays JA. Deletion of transcription factor AP-2β from the developing murine trabecular meshwork region leads to progressive glaucomatous changes. J Neurosci Res 2021; 100:638-652. [PMID: 34822722 DOI: 10.1002/jnr.24982] [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: 08/24/2021] [Revised: 09/28/2021] [Accepted: 10/03/2021] [Indexed: 11/08/2022]
Abstract
Glaucoma is one of the leading causes of irreversible blindness and can result from abnormalities in anterior segment structures required for aqueous humor outflow, including the trabecular meshwork (TM) and Schlemm's canal (SC). Transcription factors such as AP-2β play critical roles in anterior segment development. Here, we show that the Mgp-Cre knock-in (Mgp-Cre.KI) mouse can be used to target the embryonic periocular mesenchyme giving rise to the TM and SC. Fate mapping of male and female mice indicates that AP-2β loss causes a decrease in iridocorneal angle cells derived from Mgp-Cre.KI-expressing populations compared to controls. Moreover, histological analyses revealed peripheral iridocorneal adhesions in AP-2β mutants that were accompanied by a decrease in expression of TM and SC markers, as observed using immunohistochemistry. In addition, rebound tonometry showed significantly higher intraocular pressure (IOP) that was correlated with a progressive significant loss of retinal ganglion cells, reduced retinal thickness, and reduced retinal function, as measured using an electroretinogram, in AP-2β mutants compared with controls, reflecting pathology described in late-stage glaucoma patients. Importantly, elevated IOP in AP-2β mutants was significantly reduced by treatment with latanoprost, a prostaglandin analog that increases unconventional outflow. These findings demonstrate that AP-2β is critical for TM and SC development, and that these mutant mice can serve as a model for understanding and treating progressive human primary angle-closure glaucoma.
Collapse
Affiliation(s)
- Aftab Taiyab
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Monica Akula
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Japnit Dham
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Paula Deschamps
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Heather Sheardown
- Department of Chemical Engineering, McMaster University, Hamilton, ON, Canada
| | - Trevor Williams
- Department of Craniofacial Biology, University of Colorado, Aurora, CO, USA
| | - Teresa Borrás
- Department of Ophthalmology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Judith A West-Mays
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
26
|
Youkilis JC, Bassnett S. Single-cell RNA-sequencing analysis of the ciliary epithelium and contiguous tissues in the mouse eye. Exp Eye Res 2021; 213:108811. [PMID: 34717927 PMCID: PMC8860325 DOI: 10.1016/j.exer.2021.108811] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 10/08/2021] [Accepted: 10/24/2021] [Indexed: 12/12/2022]
Abstract
The ciliary epithelium plays a central role in ocular homeostasis but cells of the pigmented and non-pigmented layers are difficult to isolate physically and study. Here we used single-cell RNA-sequencing (scRNA-seq) to analyze the transcriptional signatures of cells harvested from the ciliary body and contiguous tissues. Microdissected tissue was dissociated by collagenase digestion and the transcriptomes of individual cells were obtained using a droplet-based scRNA-seq approach. In situ hybridization was used to verify the expression patterns of selected differentially-expressed genes. High quality transcriptomes were obtained from 10,024 cells and unsupervised clustering distinguished 22 cell types. Although efforts were made to specifically isolate the ciliary body, approximately half of the sequenced cells were derived from the adjacent retina. Cluster identities were assigned using expression of canonical markers or cluster-specific genes. The transcriptional signature of cells in the PCE and NPCE were distinct from each other and from cells in contiguous tissues. PCE cell transcriptomes were characterized by genes involved in melanin synthesis and transport proteins such as Slc4a4. Among the most differentially expressed genes in NPCE cells were those encoding members of the Zic family of transcription factors (Zic1, 2, 4), collagen XVIII (Col18a1), and corticotrophin-releasing hormone-binding protein (Crhbp). The ocular melanocyte population was distinguished by expression of the gap junction genes Gjb2 and Gjb6. Two fibroblast signatures were detected in the ciliary body preparation and shown by in situ hybridization to correspond to uveal and scleral populations. This cell atlas for the ciliary body and contiguous layers represents a useful resource that may facilitate studies into the development of the ciliary epithelium, the production of the aqueous and vitreous humors, and the synthesis of the ciliary zonule.
Collapse
Affiliation(s)
- J C Youkilis
- Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - S Bassnett
- Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
27
|
Sharif NA. Therapeutic Drugs and Devices for Tackling Ocular Hypertension and Glaucoma, and Need for Neuroprotection and Cytoprotective Therapies. Front Pharmacol 2021; 12:729249. [PMID: 34603044 PMCID: PMC8484316 DOI: 10.3389/fphar.2021.729249] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 08/18/2021] [Indexed: 12/11/2022] Open
Abstract
Damage to the optic nerve and the death of associated retinal ganglion cells (RGCs) by elevated intraocular pressure (IOP), also known as glaucoma, is responsible for visual impairment and blindness in millions of people worldwide. The ocular hypertension (OHT) and the deleterious mechanical forces it exerts at the back of the eye, at the level of the optic nerve head/optic disc and lamina cribosa, is the only modifiable risk factor associated with glaucoma that can be treated. The elevated IOP occurs due to the inability of accumulated aqueous humor (AQH) to egress from the anterior chamber of the eye due to occlusion of the major outflow pathway, the trabecular meshwork (TM) and Schlemm’s canal (SC). Several different classes of pharmaceutical agents, surgical techniques and implantable devices have been developed to lower and control IOP. First-line drugs to promote AQH outflow via the uveoscleral outflow pathway include FP-receptor prostaglandin (PG) agonists (e.g., latanoprost, travoprost and tafluprost) and a novel non-PG EP2-receptor agonist (omidenepag isopropyl, Eybelis®). TM/SC outflow enhancing drugs are also effective ocular hypotensive agents (e.g., rho kinase inhibitors like ripasudil and netarsudil; and latanoprostene bunod, a conjugate of a nitric oxide donor and latanoprost). One of the most effective anterior chamber AQH microshunt devices is the Preserflo® microshunt which can lower IOP down to 10–13 mmHg. Other IOP-lowering drugs and devices on the horizon will be also discussed. Additionally, since elevated IOP is only one of many risk factors for development of glaucomatous optic neuropathy, a treatise of the role of inflammatory neurodegeneration of the optic nerve and retinal ganglion cells and appropriate neuroprotective strategies to mitigate this disease will also be reviewed and discussed.
Collapse
Affiliation(s)
- Najam A Sharif
- Global Alliances and External Research, Ophthalmology Innovation Center, Santen Inc., Emeryville, CA, United States
| |
Collapse
|
28
|
Li X, Cai S, He Z, Reilly J, Zeng Z, Strang N, Shu X. Metabolomics in Retinal Diseases: An Update. BIOLOGY 2021; 10:944. [PMID: 34681043 PMCID: PMC8533136 DOI: 10.3390/biology10100944] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 09/16/2021] [Accepted: 09/18/2021] [Indexed: 12/17/2022]
Abstract
Retinal diseases are a leading cause of visual loss and blindness, affecting a significant proportion of the population worldwide and having a detrimental impact on quality of life, with consequent economic burden. The retina is highly metabolically active, and a number of retinal diseases are associated with metabolic dysfunction. To better understand the pathogenesis underlying such retinopathies, new technology has been developed to elucidate the mechanism behind retinal diseases. Metabolomics is a relatively new "omics" technology, which has developed subsequent to genomics, transcriptomics, and proteomics. This new technology can provide qualitative and quantitative information about low-molecular-weight metabolites (M.W. < 1500 Da) in a given biological system, which shed light on the physiological or pathological state of a cell or tissue sample at a particular time point. In this article we provide an extensive review of the application of metabolomics to retinal diseases, with focus on age-related macular degeneration (AMD), diabetic retinopathy (DR), retinopathy of prematurity (ROP), glaucoma, and retinitis pigmentosa (RP).
Collapse
Affiliation(s)
- Xing Li
- School of Basic Medical Sciences, Shaoyang University, Shaoyang 422000, China; (X.L.); (Z.H.)
| | - Shichang Cai
- Department of Human Anatomy, School of Medicine, Hunan University of Medicine, Huaihua 418000, China;
| | - Zhiming He
- School of Basic Medical Sciences, Shaoyang University, Shaoyang 422000, China; (X.L.); (Z.H.)
| | - James Reilly
- Department of Biological and Biomedical Sciences, Glasgow Caledonian University, Glasgow G4 0BA, UK;
| | - Zhihong Zeng
- College of Biological and Environmental Engineering, Changsha University, Changsha 410022, China;
| | - Niall Strang
- Department of Vision Science, Glasgow Caledonian University, Glasgow G4 0BA, UK;
| | - Xinhua Shu
- School of Basic Medical Sciences, Shaoyang University, Shaoyang 422000, China; (X.L.); (Z.H.)
- Department of Biological and Biomedical Sciences, Glasgow Caledonian University, Glasgow G4 0BA, UK;
- Department of Vision Science, Glasgow Caledonian University, Glasgow G4 0BA, UK;
| |
Collapse
|
29
|
Myer C, Abdelrahman L, Banerjee S, Khattri RB, Merritt ME, Junk AK, Lee RK, Bhattacharya SK. Aqueous humor metabolite profile of pseudoexfoliation glaucoma is distinctive. Mol Omics 2021; 16:425-435. [PMID: 32149291 DOI: 10.1039/c9mo00192a] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Pseudoexfoliation (PEX) is a known cause of secondary open angle glaucoma. PEX glaucoma is associated with structural and metabolic changes in the eye. Despite similarities, PEX and primary open angle glaucoma (POAG) may have differences in the composition of metabolites. We analyzed the metabolites of the aqueous humor (AH) of PEX subjects sequentially first using nuclear magnetic resonance (1H NMR: HSQC and TOCSY), and subsequently with liquid chromatography tandem mass spectrometry (LC-MS/MS) implementing isotopic ratio outlier analysis (IROA) quantification. The findings were compared with previous results for POAG and control subjects analyzed using identical sequential steps. We found significant differences in metabolites between the three conditions. Principle component analysis (PCA) and partial least squares discriminant analysis (PLS-DA) indicated clear grouping based on the metabolomes of the three conditions. We used machine learning algorithms and a percentage set of the data to train, and utilized a different or larger dataset to test whether a trained model can correctly classify the test dataset as PEX, POAG or control. Three different algorithms: linear support vector machines (SVM), deep learning, and a neural network were used for prediction. They all accurately classified the test datasets based on the AH metabolome of the sample. We next compared the AH metabolome with known AH and TM proteomes and genomes in order to understand metabolic pathways that may contribute to alterations in the AH metabolome in PEX. We found potential protein/gene pathways associated with observed significant metabolite changes in PEX.
Collapse
Affiliation(s)
- Ciara Myer
- Bascom Palmer Eye Institute, University of Miami, Miami, Florida, USA. and Miami Integrative Metabolomics Research Center, University of Miami, Miami, Florida, USA
| | - Leila Abdelrahman
- Bascom Palmer Eye Institute, University of Miami, Miami, Florida, USA. and Miami Integrative Metabolomics Research Center, University of Miami, Miami, Florida, USA
| | - Santanu Banerjee
- Bascom Palmer Eye Institute, University of Miami, Miami, Florida, USA. and Miami Integrative Metabolomics Research Center, University of Miami, Miami, Florida, USA and Department of Surgery, University of Miami, Miami, Florida, USA
| | | | | | - Anna K Junk
- Bascom Palmer Eye Institute, University of Miami, Miami, Florida, USA. and Miami Integrative Metabolomics Research Center, University of Miami, Miami, Florida, USA and Miami Veterans Affairs Healthcare System, Miami, Florida, USA
| | - Richard K Lee
- Bascom Palmer Eye Institute, University of Miami, Miami, Florida, USA. and Miami Integrative Metabolomics Research Center, University of Miami, Miami, Florida, USA
| | - Sanjoy K Bhattacharya
- Bascom Palmer Eye Institute, University of Miami, Miami, Florida, USA. and Miami Integrative Metabolomics Research Center, University of Miami, Miami, Florida, USA
| |
Collapse
|
30
|
Chung HW, Park JH, Yoo C, Kim YY. Effects of Trabecular Meshwork Width and Schlemm's Canal Area on Intraocular Pressure Reduction in Glaucoma Patients. KOREAN JOURNAL OF OPHTHALMOLOGY 2021; 35:311-317. [PMID: 34162183 PMCID: PMC8357607 DOI: 10.3341/kjo.2021.0007] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 06/18/2021] [Indexed: 11/23/2022] Open
Abstract
PURPOSE To evaluate the effects of baseline trabecular meshwork (TM) and Schlemm's canal (SC) microstructures on intraocular pressure (IOP) reduction amount in treatment-naïve patients with primary open-angle glaucoma (POAG). METHODS A total 69 eyes of POAG patients who had not been treated with IOP-lowering agent were enrolled in this retrospective study. The patients had been prescribed topical IOP-lowering agent and used it for 1 year. The morphologic features of the TM and SC were collected using anterior segment module of spectral-domain optical coherence tomography with enhanced depth imaging at baseline. Images of the nasal and temporal corneoscleral limbus were obtained with serial horizontal enhanced depth imaging B-scans and TM width and SC area were measured in each scan. We investigated the effects of baseline TM and SC microstructures on IOP reduction amount. RESULTS The baseline IOP of 69 glaucomatous eyes was 17.9 ± 3.8 mmHg, and the mean amount of IOP reduction was 3.5 ± 2.1 mmHg after 1 year. Mean TM widths of nasal and temporal sector were 470.33 ± 80.05 and 479.74 ± 79.59 μm, respectively. SC area was measured as 4,818.50 ± 1,464.28, 4,604.23 ± 1,567.73 μm2 at nasal sector and temporal sector, respectively. The correlation analysis revealed a positive correlation between SC area and average amount of IOP reduction, indicating that the larger baseline SC area, the greater the IOP drop with topical IOP-lowering agents. However, no correlation was found between TM width and IOP lowering amount in patients with POAG. CONCLUSIONS The baseline SC area showed positive correlation with the IOP reduction amount in patients with POAG. This finding suggests that the SC area can be a clinical parameter to predict the IOP reduction amount before using IOP-lowering agents in POAG patient.
Collapse
Affiliation(s)
- Hyun Woo Chung
- Department of Ophthalmology, Korea University College of Medicine, Seoul, Korea
| | - Ji-Hye Park
- Department of Ophthalmology, Korea University College of Medicine, Seoul, Korea
| | - Chungkwon Yoo
- Department of Ophthalmology, Korea University College of Medicine, Seoul, Korea
| | - Yong Yeon Kim
- Department of Ophthalmology, Korea University College of Medicine, Seoul, Korea
| |
Collapse
|
31
|
Tabak S, Schreiber-Avissar S, Beit-Yannai E. Trabecular meshwork's collagen network formation is inhibited by non-pigmented ciliary epithelium-derived extracellular vesicles. J Cell Mol Med 2021; 25:3339-3347. [PMID: 33644975 PMCID: PMC8034463 DOI: 10.1111/jcmm.16408] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 02/07/2021] [Accepted: 02/09/2021] [Indexed: 12/22/2022] Open
Abstract
The present research aims to determine whether the application of non‐pigmented ciliary epithelium cells derived extracellular vesicles to human trabecular meshwork cells affects the formation and secretion of collagen type I to the extracellular matrix formation. Following the extraction of non‐pigmented ciliary epithelium derived extracellular vesicles by a precipitation method, their size and concentration were determined using tunable resistive pulse sensing technology. Extracellular vesicles were incubated with trabecular meshwork cells for 3 days. Morphological changes of collagen type I in the extracellular matrix of trabecular meshwork cells were visualized using confocal microscopy and scanning electron microscopy. A Sirius Red assay was used to determine the total amount of collagen. Finally, collagen type I expression levels in the extracellular matrix of trabecular meshwork cells were quantified by cell western analysis. We found that non‐pigmented ciliary epithelium extracellular vesicles were very effective at preventing collagen fibres formation by the trabecular meshwork cells, and their secretion to the extracellular matrix was significantly reduced (P < .001). Morphological changes in the extracellular matrix of trabecular meshwork cells were observed. Our study indicates that non‐pigmented ciliary epithelium extracellular vesicles can be used to control collagen type I fibrillogenesis in trabecular meshwork cells. These fibrils net‐like structure is responsible for remodelling the extracellular matrix. Moreover, we suggest that targeting collagen type I fibril assembly may be a viable treatment for primary open‐angle glaucoma abnormal matrix deposition of the extracellular matrix.
Collapse
Affiliation(s)
- Saray Tabak
- Clinical Biochemistry and Pharmacology Department, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Sofia Schreiber-Avissar
- Clinical Biochemistry and Pharmacology Department, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Elie Beit-Yannai
- Clinical Biochemistry and Pharmacology Department, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| |
Collapse
|
32
|
Ishii K, Asaoka R, Omoto T, Mitaki S, Fujino Y, Murata H, Onoda K, Nagai A, Yamaguchi S, Obana A, Tanito M. Predicting intraocular pressure using systemic variables or fundus photography with deep learning in a health examination cohort. Sci Rep 2021; 11:3687. [PMID: 33574359 PMCID: PMC7878799 DOI: 10.1038/s41598-020-80839-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 12/21/2020] [Indexed: 12/17/2022] Open
Abstract
The purpose of the current study was to predict intraocular pressure (IOP) using color fundus photography with a deep learning (DL) model, or, systemic variables with a multivariate linear regression model (MLM), along with least absolute shrinkage and selection operator regression (LASSO), support vector machine (SVM), and Random Forest: (RF). Training dataset included 3883 examinations from 3883 eyes of 1945 subjects and testing dataset 289 examinations from 289 eyes from 146 subjects. With the training dataset, MLM was constructed to predict IOP using 35 systemic variables and 25 blood measurements. A DL model was developed to predict IOP from color fundus photographs. The prediction accuracy of each model was evaluated through the absolute error and the marginal R-squared (mR2), using the testing dataset. The mean absolute error with MLM was 2.29 mmHg, which was significantly smaller than that with DL (2.70 dB). The mR2 with MLM was 0.15, whereas that with DL was 0.0066. The mean absolute error (between 2.24 and 2.30 mmHg) and mR2 (between 0.11 and 0.15) with LASSO, SVM and RF were similar to or poorer than MLM. A DL model to predict IOP using color fundus photography proved far less accurate than MLM using systemic variables.
Collapse
Affiliation(s)
- Kaori Ishii
- Department of Ophthalmology, Seirei Hamamatsu General Hospital, Hamamatsu, Shizuoka, Japan
| | - Ryo Asaoka
- Department of Ophthalmology, Seirei Hamamatsu General Hospital, Hamamatsu, Shizuoka, Japan.
- Seirei Christopher University, Hamamatsu, Shizuoka, Japan.
- Department of Ophthalmology, The University of Tokyo, Tokyo, Japan.
| | - Takashi Omoto
- Department of Ophthalmology, The University of Tokyo, Tokyo, Japan
| | - Shingo Mitaki
- Department of Neurology, Shimane University Faculty of Medicine, Izumo, Japan
| | - Yuri Fujino
- Department of Ophthalmology, Seirei Hamamatsu General Hospital, Hamamatsu, Shizuoka, Japan
- Department of Ophthalmology, Shimane University Faculty of Medicine, Izumo, Japan
| | - Hiroshi Murata
- Department of Ophthalmology, The University of Tokyo, Tokyo, Japan
| | - Keiichi Onoda
- Department of Neurology, Shimane University Faculty of Medicine, Izumo, Japan
- Faculty of Psychology, Outemon Gakuin University, Osaka, Japan
| | - Atsushi Nagai
- Department of Neurology, Shimane University Faculty of Medicine, Izumo, Japan
| | - Shuhei Yamaguchi
- Department of Neurology, Shimane University Faculty of Medicine, Izumo, Japan
| | - Akira Obana
- Department of Ophthalmology, Seirei Hamamatsu General Hospital, Hamamatsu, Shizuoka, Japan
- Hamamatsu BioPhotonics Innovation Chair, Institute for Medical Photonics Research, Preeminent Medical Photonics Education & Research Center, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Masaki Tanito
- Department of Ophthalmology, Shimane University Faculty of Medicine, Izumo, Japan
| |
Collapse
|
33
|
Spinozzi E, Baldassarri C, Acquaticci L, Del Bello F, Grifantini M, Cappellacci L, Riccardo P. Adenosine receptors as promising targets for the management of ocular diseases. Med Chem Res 2021; 30:353-370. [PMID: 33519168 PMCID: PMC7829661 DOI: 10.1007/s00044-021-02704-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 12/01/2020] [Indexed: 12/15/2022]
Abstract
The ocular drug discovery arena has undergone a significant improvement in the last few years culminating in the FDA approvals of 8 new drugs. However, despite a large number of drugs, generics, and combination products available, it remains an urgent need to find breakthrough strategies and therapies for tackling ocular diseases. Targeting the adenosinergic system may represent an innovative strategy for discovering new ocular therapeutics. This review focused on the recent advance in the field and described the numerous nucleoside and non-nucleoside modulators of the four adenosine receptors (ARs) used as potential tools or clinical drug candidates.
Collapse
Affiliation(s)
- Eleonora Spinozzi
- School of Pharmacy Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, 62032 Camerino, Italy
| | - Cecilia Baldassarri
- School of Pharmacy Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, 62032 Camerino, Italy
| | - Laura Acquaticci
- School of Pharmacy Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, 62032 Camerino, Italy
| | - Fabio Del Bello
- School of Pharmacy Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, 62032 Camerino, Italy
| | - Mario Grifantini
- School of Pharmacy Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, 62032 Camerino, Italy
| | - Loredana Cappellacci
- School of Pharmacy Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, 62032 Camerino, Italy
| | - Petrelli Riccardo
- School of Pharmacy Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, 62032 Camerino, Italy
| |
Collapse
|
34
|
Zhu W, Hou F, Fang J, Bahrani Fard MR, Liu Y, Ren S, Wu S, Qi Y, Sui S, Read AT, Sherwood JM, Zou W, Yu H, Zhang J, Overby DR, Wang N, Ethier CR, Wang K. The role of Piezo1 in conventional aqueous humor outflow dynamics. iScience 2021; 24:102042. [PMID: 33532718 PMCID: PMC7829208 DOI: 10.1016/j.isci.2021.102042] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 12/16/2020] [Accepted: 01/05/2021] [Indexed: 12/16/2022] Open
Abstract
Controlling intraocular pressure (IOP) remains the mainstay of glaucoma therapy. The trabecular meshwork (TM), the key tissue responsible for aqueous humor (AH) outflow and IOP maintenance, is very sensitive to mechanical forces. However, it is not understood whether Piezo channels, very sensitive mechanosensors, functionally influence AH outflow. Here, we characterize the role of Piezo1 in conventional AH outflow. Immunostaining and western blot analysis showed that Piezo1 is widely expressed by TM. Patch-clamp recordings in TM cells confirmed the activation of Piezo1-derived mechanosensitive currents. Importantly, the antagonist GsMTx4 for mechanosensitive channels significantly decreased steady-state facility, yet activation of Piezo1 by the specific agonist Yoda1 did not lead to a facility change. Furthermore, GsMTx4, but not Yoda1, caused a significant increase in ocular compliance, a measure of the eye's transient response to IOP perturbation. Our findings demonstrate a potential role for Piezo1 in conventional outflow, likely under pathological and rapid transient conditions. Piezo1 is functionally expressed in the TM, the most important tissue controlling IOP Suppression of mechanosensitive channel leads to a significant decrease in facility Our data suggest a role for Piezo in pathological situations and rapid IOP transients
Collapse
Affiliation(s)
- Wei Zhu
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao University Medical College, 38 Dengzhou Road, Qingdao 266021, Shandong, China.,Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University & Capital Medical University, Beijing 100730, China
| | - Fei Hou
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao University Medical College, 38 Dengzhou Road, Qingdao 266021, Shandong, China.,Department of Pharmacy, Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Jingwang Fang
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao University Medical College, 38 Dengzhou Road, Qingdao 266021, Shandong, China
| | - Mohammad Reza Bahrani Fard
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta 30332, GA, USA
| | - Yani Liu
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao University Medical College, 38 Dengzhou Road, Qingdao 266021, Shandong, China
| | - Shouyan Ren
- Department of Pharmacy, Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Shen Wu
- Beijing Institute of Ophthalmology, Beijing Tongren Hospital Eye Center, Beijing 100730 China
| | - Yunkun Qi
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao University Medical College, 38 Dengzhou Road, Qingdao 266021, Shandong, China
| | - Shangru Sui
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao University Medical College, 38 Dengzhou Road, Qingdao 266021, Shandong, China
| | - A Thomas Read
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta 30332, GA, USA
| | | | - Wei Zou
- School of Mechatronic Engineering, Jiangsu Normal University, Xuzhou 221116, China
| | - Hongxia Yu
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao University Medical College, 38 Dengzhou Road, Qingdao 266021, Shandong, China
| | - Jingxue Zhang
- Beijing Institute of Ophthalmology, Beijing Tongren Hospital Eye Center, Beijing 100730 China
| | - Darryl R Overby
- Department of Bioengineering, Imperial College London, London, UK
| | - Ningli Wang
- Beijing Institute of Ophthalmology, Beijing Tongren Hospital Eye Center, Beijing 100730 China
| | - C Ross Ethier
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta 30332, GA, USA.,Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, 30332, GA, United States
| | - KeWei Wang
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao University Medical College, 38 Dengzhou Road, Qingdao 266021, Shandong, China.,Institute of Innovative Drugs, Qingdao University, 38 Dengzhou Road, Qingdao 266021, Shandong, China
| |
Collapse
|
35
|
Vernazza S, Tirendi S, Bassi AM, Traverso CE, Saccà SC. Neuroinflammation in Primary Open-Angle Glaucoma. J Clin Med 2020; 9:E3172. [PMID: 33007927 PMCID: PMC7601106 DOI: 10.3390/jcm9103172] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/24/2020] [Accepted: 09/28/2020] [Indexed: 12/18/2022] Open
Abstract
Primary open-angle glaucoma (POAG) is the second leading cause of irreversible blindness worldwide. Increasing evidence suggests oxidative damage and immune response defects are key factors contributing to glaucoma onset. Indeed, both the failure of the trabecular meshwork tissue in the conventional outflow pathway and the neuroinflammation process, which drives the neurodegeneration, seem to be linked to the age-related over-production of free radicals (i.e., mitochondrial dysfunction) and to oxidative stress-linked immunostimulatory signaling. Several previous studies have described a wide range of oxidative stress-related makers which are found in glaucomatous patients, including low levels of antioxidant defences, dysfunction/activation of glial cells, the activation of the NF-κB pathway and the up-regulation of pro-inflammatory cytokines, and so on. However, the intraocular pressure is still currently the only risk factor modifiable by medication or glaucoma surgery. This present review aims to summarize the multiple cellular processes, which promote different risk factors in glaucoma including aging, oxidative stress, trabecular meshwork defects, glial activation response, neurodegenerative insults, and the altered regulation of immune response.
Collapse
Affiliation(s)
| | - Sara Tirendi
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genoa, Italy; (S.T.); (A.M.B.)
- Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research (Centro 3R), Italy
| | - Anna Maria Bassi
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genoa, Italy; (S.T.); (A.M.B.)
- Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research (Centro 3R), Italy
| | - Carlo Enrico Traverso
- Clinica Oculistica, DiNOGMI, University of Genoa, 16132 Genoa, Italy;
- Ophthalmology Unit, IRCCS-Polyclinic San Martino Hospital, 16132 Genoa, Italy;
| | | |
Collapse
|
36
|
Liu Q, Liu D, Yan D, Huang W, Ji X, Hui J, Tang Z. Gender-Specific Association Between Serum Uric Acid and Incident High Intraocular Pressure in Chinese Population: A Cross-Sectional Study. Invest Ophthalmol Vis Sci 2020; 61:10. [PMID: 32897376 PMCID: PMC7488210 DOI: 10.1167/iovs.61.11.10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Purpose The purpose of this study was to investigate the relationship between high intraocular pressure (IOP) and uric acid. Methods In a retrospective cross-sectional study, 19,147 participants were included in 2018. Serum uric acid (SUA) was cut to four groups as Q1 to Q4, according to the quartiles. The odds ratio (OR) and 95% confidence interval (CI) of different SUA levels were estimated by a binomial logistic regression model in men and women. A restrictive cubic spline method was used to estimate the dose-response relationship between uric acid and high IOP. Subgroup analysis was performed to find the gender-specific association between uric acid and high IOP. Results In women, after adjusting for confounding factors, the Q3 and Q4 of SUA levels were significantly associated with the risk of high IOP. The OR with 95% CI for Q3 and Q4 were 1.77 (1.22, 2.57) and 1.51 (1.01, 2.26), respectively, Q1 as a reference. For men, SUA levels were not associated with the incidence of high IOP. Moreover, the spline analysis found an inverted U‐shaped relationship between uric acid and high IOP in women (P = 0.0171). Conclusions Elevated levels of SUAwere independently associated with an increased risk of high IOP in women, but not in men. In addition, uric acid had an inverse U-shaped nonlinear dose-response relationship with high IOP in women.
Collapse
Affiliation(s)
- Qianqian Liu
- Department of Biostatistics, School of Public Health, Medical College of Soochow University, Suzhou, China.,Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
| | - Dan Liu
- Health Management Center, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Derui Yan
- Department of Biostatistics, School of Public Health, Medical College of Soochow University, Suzhou, China.,Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
| | - Weicun Huang
- Department of Biostatistics, School of Public Health, Medical College of Soochow University, Suzhou, China.,Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
| | - Xiaodong Ji
- Health Management Center, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jie Hui
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zaixiang Tang
- Department of Biostatistics, School of Public Health, Medical College of Soochow University, Suzhou, China.,Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
| |
Collapse
|
37
|
Holappa M, Vapaatalo H, Vaajanen A. Local ocular renin-angiotensin-aldosterone system: any connection with intraocular pressure? A comprehensive review. Ann Med 2020; 52:191-206. [PMID: 32308046 PMCID: PMC7877937 DOI: 10.1080/07853890.2020.1758341] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 04/15/2020] [Indexed: 12/28/2022] Open
Abstract
The renin-angiotensin system (RAS) is one of the oldest and most extensively studied human peptide cascades, well-known for its role in regulating blood pressure. When aldosterone is included, RAAS is involved also in fluid and electrolyte homeostasis. There are two main axes of RAAS: (1) Angiotensin (1-7), angiotensin converting enzyme 2 and Mas receptor (ACE2-Ang(1-7)-MasR), (2) Angiotensin II, angiotensin converting enzyme 1 and angiotensin II type 1 receptor (ACE1-AngII-AT1R). In its entirety, RAAS comprises dozens of angiotensin peptides, peptidases and seven receptors. The first mentioned axis is known to counterbalance the deleterious effects of the latter axis. In addition to the systemic RAAS, tissue-specific regulatory systems have been described in various organs, evidence that RAAS is both an endocrine and an autocrine system. These local regulatory systems, such as the one present in the vascular endothelium, are responsible for long-term regional changes. A local RAAS and its components have been detected in many structures of the human eye. This review focuses on the local ocular RAAS in the anterior part of the eye, its possible role in aqueous humour dynamics and intraocular pressure as well as RAAS as a potential target for anti-glaucomatous drugs.KEY MESSAGESComponents of renin-angiotensin-aldosterone system have been detected in different structures of the human eye, introducing the concept of a local intraocular renin-angiotensin-aldosterone system (RAAS).Evidence is accumulating that the local ocular RAAS is involved in aqueous humour dynamics, regulation of intraocular pressure, neuroprotection and ocular pathology making components of RAAS attractive candidates when developing new effective ways to treat glaucoma.
Collapse
Affiliation(s)
- Mervi Holappa
- Medical Faculty, Department of Pharmacology, University of Helsinki, Helsinki, Finland
| | - Heikki Vapaatalo
- Medical Faculty, Department of Pharmacology, University of Helsinki, Helsinki, Finland
| | - Anu Vaajanen
- Department of Ophthalmology, Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
38
|
Scuderi L, Davinelli S, Iodice CM, Bartollino S, Scapagnini G, Costagliola C, Scuderi G. Melatonin: Implications for Ocular Disease and Therapeutic Potential. Curr Pharm Des 2020; 25:4185-4191. [PMID: 31724508 DOI: 10.2174/1381612825666191113110225] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 11/12/2019] [Indexed: 02/08/2023]
Abstract
Melatonin, an indoleamine secreted mainly by the pineal gland, is known to modulate a wide range of circadian functions. However, this neurohormone is also synthesized within the eye and acts directly on ocular structures to mediate a variety of physiological processes. This review is focused on the role and therapeutic potential of melatonin in ocular diseases. We summarize data indicating that melatonin may represent a powerful tool to counteract ocular dysfunctions such as uveitis, glaucoma, age-related macular degeneration, and diabetic retinopathy. A search strategy was conducted to identify studies in PubMed (January 1990 to September 2017). In particular, we included experimental studies, clinical trials, and reviews to provide suitable insights and elucidations regarding the action of melatonin on age-related ocular disorders. Literature data suggest that melatonin could potentially protect ocular tissues by decreasing the production of free radicals and pro-inflammatory mediators. Additionally, melatonin appears to be safe and well-tolerated, even at high doses, and no adverse/side effects were reported. Although this topic remains under intense investigation, we can conclude that melatonin, as a single agent or in combination with other drugs, is an attractive pharmacological candidate for age-related ocular diseases.
Collapse
Affiliation(s)
- Luca Scuderi
- Neuroscience, Mental Health and Sense Organs Department, Faculty of Medicine and Psychology, University of Rome "La Sapienza", Rome, Italy
| | - Sergio Davinelli
- Department of Medicine and Health Sciences "V. Tiberio", University of Molise, Campobasso, Italy
| | - Clemente Maria Iodice
- Neuroscience, Mental Health and Sense Organs Department, Faculty of Medicine and Psychology, University of Rome "La Sapienza", Rome, Italy
| | - Silvia Bartollino
- Department of Medicine and Health Sciences "V. Tiberio", University of Molise, Campobasso, Italy
| | - Giovanni Scapagnini
- Department of Medicine and Health Sciences "V. Tiberio", University of Molise, Campobasso, Italy
| | - Ciro Costagliola
- Department of Medicine and Health Sciences "V. Tiberio", University of Molise, Campobasso, Italy
| | - Gianluca Scuderi
- Neuroscience, Mental Health and Sense Organs Department, Faculty of Medicine and Psychology, University of Rome "La Sapienza", Rome, Italy
| |
Collapse
|
39
|
Lu R, Soden PA, Lee E. Tissue-Engineered Models for Glaucoma Research. MICROMACHINES 2020; 11:mi11060612. [PMID: 32599818 PMCID: PMC7345325 DOI: 10.3390/mi11060612] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 06/21/2020] [Accepted: 06/22/2020] [Indexed: 12/20/2022]
Abstract
Glaucoma is a group of optic neuropathies characterized by the progressive degeneration of retinal ganglion cells (RGCs). Patients with glaucoma generally experience elevations in intraocular pressure (IOP), followed by RGC death, peripheral vision loss and eventually blindness. However, despite the substantial economic and health-related impact of glaucoma-related morbidity worldwide, the surgical and pharmacological management of glaucoma is still limited to maintaining IOP within a normal range. This is in large part because the underlying molecular and biophysical mechanisms by which glaucomatous changes occur are still unclear. In the present review article, we describe current tissue-engineered models of the intraocular space that aim to advance the state of glaucoma research. Specifically, we critically evaluate and compare both 2D and 3D-culture models of the trabecular meshwork and nerve fiber layer, both of which are key players in glaucoma pathophysiology. Finally, we point out the need for novel organ-on-a-chip models of glaucoma that functionally integrate currently available 3D models of the retina and the trabecular outflow pathway.
Collapse
Affiliation(s)
- Renhao Lu
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA;
| | - Paul A. Soden
- College of Human Ecology, Cornell University, Ithaca, NY 14853, USA;
| | - Esak Lee
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA;
- Correspondence: ; Tel.: +1-607-255-8491
| |
Collapse
|
40
|
Systemic factors associated with intraocular pressure among subjects in a health examination program in Japan. PLoS One 2020; 15:e0234042. [PMID: 32492062 PMCID: PMC7269229 DOI: 10.1371/journal.pone.0234042] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Accepted: 05/18/2020] [Indexed: 02/06/2023] Open
Abstract
Background To elucidate the possible effect of various systemic factors on intraocular pressure (IOP) using a dataset from a health examination program database in Japan. Methods This cross-sectional study included 1569 subjects selected from the 2287 subjects who comprised the database. Various systemic parameters including age, sex, height, body weight, waist circumference, percent body fat, blood pressure (BP), pulse rate, body mass index, 28 blood examination values, intimal medial thicknesses of both carotid arteries, and intraocular pressure (IOP) values measured by non-contact tonometry in both eyes were collected. The possible correlation between the IOP and other parameters was assessed initially by univariate analyses followed by multivariate analyses. Results Stepwise multivariate analyses, which included all parameters extracted by the univariate analyses (p<0.1) and sex, identified the same six parameters as indicators of the IOP values for each right and left IOP model. Among the parameters, age (r = -0.05 and -0.04/year for right and left IOPs, respectively) was associated negatively and the percent body fat (r = 0.06 and 0.05/%), systolic BP (r = 0.02 and 0.03/mmHg), pulse rate (r = 0.03 and 0.03/counts/minutes), albumin (r = 1.12 and 1.00/g/dL), and hemoglobin A1c (r = 0.38 and 0.44/%) were associated positively with the IOP in each eye. Conclusions Older age was associated with low IOP, while factors reflecting the metabolic syndrome were associated with high IOP in our study population.
Collapse
|
41
|
Myer C, Perez J, Abdelrahman L, Mendez R, Khattri RB, Junk AK, Bhattacharya SK. Differentiation of soluble aqueous humor metabolites in primary open angle glaucoma and controls. Exp Eye Res 2020; 194:108024. [PMID: 32246983 DOI: 10.1016/j.exer.2020.108024] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 03/22/2020] [Accepted: 03/23/2020] [Indexed: 01/06/2023]
Abstract
We report an analysis of the aqueous humor (AH) metabolome of primary open angle glaucoma (POAG) in comparison to normal controls. The AH samples were obtained from human donors [control (n = 35), POAG (n = 23)]. The AH samples were subjected to one-dimensional 1H nuclear magnetic resonance (NMR) analyses on a Bruker Avance 600 MHz instrument with a 1.7 mM NMR probe. The same samples were then subjected to isotopic ratio outlier analysis (IROA) using a Q Exactive orbitrap mass spectrometer after chromatography on an Accela 600 HPLC. Clusterfinder Build 3.1.10 was used for identification and quantification based on long-term metabolite matrix standards. In total, 278 metabolites were identified in control samples and 273 in POAG AH. The metabolites identified were fed into previously reported proteome and genome information and the OmicsNet interaction network generator to construct a protein-metabolite interactions network with an embedded protein-protein network. Significant differences in metabolite composition in POAG compared to controls were identified indicating potential protein/gene pathways associated with these metabolites. These results will expand our previous understanding of the impeded AH metabolite composition, provide new insight into the regulation of AH outflow, and likely aid in future AH and trabecular meshwork multi-omics network analyses.
Collapse
Affiliation(s)
- Ciara Myer
- Bascom Palmer Eye Institute, University of Miami, Miami, FL, USA; Miami Integrative Metabolomics Research Center, University of Miami, Miami, FL, USA
| | - Jordan Perez
- Bascom Palmer Eye Institute, University of Miami, Miami, FL, USA; Miami Integrative Metabolomics Research Center, University of Miami, Miami, FL, USA; Case Western Reserve University, Cleveland, OH, USA
| | - Leila Abdelrahman
- Bascom Palmer Eye Institute, University of Miami, Miami, FL, USA; Miami Integrative Metabolomics Research Center, University of Miami, Miami, FL, USA
| | - Roberto Mendez
- Bascom Palmer Eye Institute, University of Miami, Miami, FL, USA; Miami Integrative Metabolomics Research Center, University of Miami, Miami, FL, USA; Department of Surgery, University of Miami, Miami, FL, USA
| | - Ram B Khattri
- Department of Biochemistry and Molecular Biochemistry, University of Florida, Gainesville, FL, USA
| | - Anna K Junk
- Bascom Palmer Eye Institute, University of Miami, Miami, FL, USA; Miami Integrative Metabolomics Research Center, University of Miami, Miami, FL, USA; Miami Veterans Affairs Health Care System, Miami, FL, USA
| | - Sanjoy K Bhattacharya
- Bascom Palmer Eye Institute, University of Miami, Miami, FL, USA; Miami Integrative Metabolomics Research Center, University of Miami, Miami, FL, USA.
| |
Collapse
|
42
|
Ghaffari Sharaf M, Damji KF, Unsworth LD. Recent advances in risk factors associated with ocular exfoliation syndrome. Acta Ophthalmol 2020; 98:113-120. [PMID: 31736276 DOI: 10.1111/aos.14298] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 10/13/2019] [Indexed: 12/12/2022]
Abstract
Exfoliation syndrome is generally considered a progressive age-related systemic disorder of the extracellular matrix, which is clinically characterized through the observation of flaky white aggregates on ocular tissues. Exfoliation syndrome is directly linked to exfoliative glaucoma in elderly patients, where it is known as the most common identifiable cause of open-angle glaucoma. Despite the identification of various risk factors associated with exfoliation syndrome, the exact pathogenesis of this syndrome has not been fully elucidated. There is a growing number of genome-wide association studies in different populations around the world to identify genetic factors underlying exfoliation syndrome. Besides variants in LOXL1 and CACNA1A genes, new loci have been recently identified which are believed to be associated with exfoliation syndrome. Among different genetic factors, functional variants might help to better understand mechanisms underlying this systemic disorder. Besides genetic factors, epigenetic regulation of different gene expression patterns has been thought to play a role in its pathogenesis. Other factors have been also considered to be involved in the development of exfoliation syndrome at cellular organelles level where mitochondrial impairment and autophagy dysfunction have been suggested in relation to exfoliation syndrome. This review addresses the most recent findings on genetic factors as well as cellular and molecular mechanisms involved in both the development and progression of exfoliation syndrome.
Collapse
Affiliation(s)
- Mehdi Ghaffari Sharaf
- Department of Chemical and Materials Engineering University of Alberta Edmonton Alberta Canada
| | - Karim F. Damji
- Department of Ophthalmology and Visual Sciences University of Alberta Edmonton Alberta Canada
| | - Larry D. Unsworth
- Department of Chemical and Materials Engineering University of Alberta Edmonton Alberta Canada
| |
Collapse
|
43
|
Shin DY, Jung KI, Park HYL, Park CK. Risk Factors for Choroidal Detachment After Ahmed Valve Implantation in Glaucoma Patients. Am J Ophthalmol 2020; 211:105-113. [PMID: 31730841 DOI: 10.1016/j.ajo.2019.10.035] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 10/24/2019] [Accepted: 10/25/2019] [Indexed: 12/29/2022]
Abstract
PURPOSE To assess the incidence and risk factors related to choroidal detachment after glaucoma drainage device (Ahmed valve) implantation. DESIGN Retrospective case-control study. METHODS A total of 188 eyes of 188 glaucoma patients were enrolled who underwent Ahmed valve implantation surgery. Patients were divided into 2 groups according to the presence or absence of choroidal detachment. The data were analyzed for factors associated with choroidal detachment. Separately, we divided eyes with choroidal detachment into 2 subgroups according to severity and conducted a subanalysis. In addition, we also analyzed the factors associated with chamber collapse. RESULTS The incidence of choroidal detachment was 35.1% according to wide-field fundus photography and 16.9% according to 45-degree fundus photography. The current study showed that age, central corneal thickness, axial length, etiology of glaucoma, history of cataract or glaucoma, hypertension, diabetes, and severity of the visual field (MD) were different between the choroidal detachment and nonchoroidal detachment groups. A multivariate analysis showed significant differences in age (P = .035), etiology of glaucoma (pseudoexfoliation; PEX) (P = .028), lens status (pseudophakia) (P = .011), and hypertension (P = .011). The greater the intraocular pressure difference before and after surgery, the greater the size of the choroidal detachment. Chamber collapse risk was associated with only short axial length. CONCLUSION The detection of choroidal detachment after Ahmed valve implantation can be increased according to the introduction of wide fundus photography. The risk of choroidal detachment is associated with the etiology of glaucoma (PEX), older age, pseudophakia (lens status), and hypertension.
Collapse
|
44
|
A Theoretical Approach for the Electrochemical Characterization of Ciliary Epithelium. Life (Basel) 2020; 10:life10020008. [PMID: 31979304 PMCID: PMC7175328 DOI: 10.3390/life10020008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 01/19/2020] [Accepted: 01/19/2020] [Indexed: 02/07/2023] Open
Abstract
The ciliary epithelium (CE) is the primary site of aqueous humor (AH) production, which results from the combined action of ultrafiltration and ionic secretion. Modulation of ionic secretion is a fundamental target for drug therapy in glaucoma, and therefore it is important to identify the main factors contributing to it. As several ion transporters have been hypothesized as relevant players in CE physiology, we propose a theoretical approach to complement experimental methods in characterizing their role in the electrochemical and fluid-dynamical conditions of CE. As a first step, we compare two model configurations that differ by (i) types of transporters included for ion exchange across the epithelial membrane, and by (i) presence or absence of the intracellular production of carbonic acid mediated by the carbonic anhydrase enzyme. The proposed model configurations do not include neurohumoral mechanisms such as P2Y receptor-dependent, cAMP, or calcium-dependent pathways, which occur in the ciliary epithelium bilayer and influence the activity of ion transporters, pumps, and channels present in the cell membrane. Results suggest that one of the two configurations predicts sodium and potassium intracellular concentrations and transmembrane potential much more accurately than the other. Because of its quantitative prediction power, the proposed theoretical approach may help relate phenomena at the cellular scale, that cannot be accessed clinically, with phenomena occurring at the scale of the whole eye, for which clinical assessment is feasible.
Collapse
|
45
|
Reyes-Resina I, Awad Alkozi H, del Ser-Badia A, Sánchez-Naves J, Lillo J, Jiménez J, Pintor J, Navarro G, Franco R. Expression of Melatonin and Dopamine D 3 Receptor Heteromers in Eye Ciliary Body Epithelial Cells and Negative Correlation with Ocular Hypertension. Cells 2020; 9:cells9010152. [PMID: 31936298 PMCID: PMC7016594 DOI: 10.3390/cells9010152] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 12/23/2019] [Accepted: 01/02/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Experiments in the late nineties showed an inverse relationship in the eye levels of melatonin and dopamine, thereby constituting an example of eye parameters that are prone to circadian variations. The underlying mechanisms are not known but these relevant molecules act via specific cell surface dopamine and melatonin receptors. This study investigated whether these receptors formed heteromers whose function impact on eye physiology. We performed biophysical assays to identify interactions in heterologous systems. Particular heteromer functionality was detected using Gi coupling, MAPK activation, and label-free assays. The expression of the heteroreceptor complexes was assessed using proximity ligation assays in cells producing the aqueous humor and human eye samples. Dopamine D3 receptors (D3Rs) were identified in eye ciliary body epithelial cells. We discovered heteromers formed by D3R and either MT1 (MT1R) or MT2 (MT2R) melatonin receptors. Heteromerization led to the blockade of D3R-Gi coupling and regulation of signaling to the MAPK pathway. Heteromer expression was negatively correlated with intraocular hypertension. CONCLUSIONS Heteromers likely mediate melatonin and dopamine actions in structures regulating intraocular pressure. Significant expression of D3R-MT1R and D3R-MT1R was associated with normotensive conditions, whereas expression diminished in a cell model of hypertension. A clear trend of expression reduction was observed in samples from glaucoma cases. The trend was marked but no statistical analysis was possible as the number of available eyes was 2.
Collapse
Affiliation(s)
- Irene Reyes-Resina
- Department of Biochemistry and Molecular Biomedicine, School of Biology, Universitat de Barcelona, 08028 Barcelona, Spain;
- Neuroplasticity Research Group, Leibniz Institute for Neurobiology, 39118 Magdeburg, Germany
- Centro de Investigación en Red, Enfermedades Neurodegenerativas, CiberNed, Instituto de Salud Carlos III, 28029 Madrid, Spain; (A.d.S.-B.); (J.J.)
- Correspondence: (I.R.-R.); (G.N.); or (R.F.); Tel.: +34-934021208 (I.R.-R. & G.N.)
| | - Hanan Awad Alkozi
- Department of Biochemistry and Molecular Biology, Faculty of Optics and Optometry, University Complutense of Madrid, 28037 Madrid, Spain; (H.A.A.); (J.P.)
| | - Anna del Ser-Badia
- Centro de Investigación en Red, Enfermedades Neurodegenerativas, CiberNed, Instituto de Salud Carlos III, 28029 Madrid, Spain; (A.d.S.-B.); (J.J.)
- Department de Bioquímica i Biologia Molecular, Institut de Neurociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, 08193 Barcelona, Spain
| | - Juan Sánchez-Naves
- Department of Ophthalmology, Balearic Islands Institute of Ophthalmology, 07013 Palma de Mallorca, Mallorca, Spain;
| | - Jaume Lillo
- Department of Biochemistry and Molecular Biomedicine, School of Biology, Universitat de Barcelona, 08028 Barcelona, Spain;
- Centro de Investigación en Red, Enfermedades Neurodegenerativas, CiberNed, Instituto de Salud Carlos III, 28029 Madrid, Spain; (A.d.S.-B.); (J.J.)
| | - Jasmina Jiménez
- Centro de Investigación en Red, Enfermedades Neurodegenerativas, CiberNed, Instituto de Salud Carlos III, 28029 Madrid, Spain; (A.d.S.-B.); (J.J.)
| | - Jesús Pintor
- Department of Biochemistry and Molecular Biology, Faculty of Optics and Optometry, University Complutense of Madrid, 28037 Madrid, Spain; (H.A.A.); (J.P.)
| | - Gemma Navarro
- Centro de Investigación en Red, Enfermedades Neurodegenerativas, CiberNed, Instituto de Salud Carlos III, 28029 Madrid, Spain; (A.d.S.-B.); (J.J.)
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Universitat de Barcelona, 08027 Barcelona, Spain
- Correspondence: (I.R.-R.); (G.N.); or (R.F.); Tel.: +34-934021208 (I.R.-R. & G.N.)
| | - Rafael Franco
- Centro de Investigación en Red, Enfermedades Neurodegenerativas, CiberNed, Instituto de Salud Carlos III, 28029 Madrid, Spain; (A.d.S.-B.); (J.J.)
- School of Chemistry, Universitat de Barcelona, 08028 Barcelona, Spain
- Correspondence: (I.R.-R.); (G.N.); or (R.F.); Tel.: +34-934021208 (I.R.-R. & G.N.)
| |
Collapse
|
46
|
Hertle R, Dell'Osso L, Jacobs J, Yang D, Dumire J, Evano-Chapman M. Topical lambda-cyhalothrin in reducing eye oscillations in a canine model of infantile nystagmus syndrome. Indian J Ophthalmol 2020; 68:2190-2195. [PMID: 32971638 PMCID: PMC7728007 DOI: 10.4103/ijo.ijo_586_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
47
|
Chen YJ, Chen JT, Tai MC, Liang CM, Chen YY, Kao TW, Fang WH, Chen WL. Examining the associations among intraocular pressure, hepatic steatosis, and anthropometric parameters. Medicine (Baltimore) 2019; 98:e17598. [PMID: 31651867 PMCID: PMC6824641 DOI: 10.1097/md.0000000000017598] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 08/15/2019] [Accepted: 09/19/2019] [Indexed: 01/28/2023] Open
Abstract
Emerging evidences had reported the positive relationship between obesity and intraocular pressure (IOP). The aim of the present study was to investigate the association between hepatic steatosis and IOP in an adult Taiwanese population.Seven thousand seven hundred twelve males and 6325 females who received a health examination at the Tri-Service General Hospital during the period from 2010 to 2016 were included in this study.IOP was measured by noncontact tonometry. Hepatic steatosis was diagnosed by abdominal ultrasound examination. Multivariate regression analyses were used to assess the associations among various anthropometric parameters and IOP.After adjusting for pertinent covariables, hepatic steatosis had a closer association with increased IOP than percentage body fat, body mass index, or waist circumference (β = 0.017, 95% confidence interval [CI] = 0.006, 0.028). This relationship remained significant among males in the study population (β = 0.015, 95% CI = 0.001, 0.029). Furthermore, hepatic steatosis was significantly correlated with increased risk of high IOP (odd ratios = 1.235, 95% CI = 1.041-1.465).Our study highlights that hepatic steatosis is a better index for assessing the relationship with increased IOP than other anthropometric parameters. Underlying pathophysiological mechanisms regulating the association between hepatic steatosis and increasing IOP and even the risk of glaucoma should be examined in further studies.
Collapse
Affiliation(s)
- Ying-Jen Chen
- Department of Ophthalmology, Tri-Service General Hospital
| | | | - Ming-Cheng Tai
- Department of Ophthalmology, Tri-Service General Hospital
| | | | - Yuan-Yuei Chen
- Department of Internal Medicine, Tri-Service General Hospital Songshan Branch
- Division of Family Medicine
| | - Tung-Wei Kao
- Division of Family Medicine
- Division of Geriatric Medicine, Department of Family and Community Medicine, Tri-Service General Hospital, and School of Medicine, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | | | - Wei-Liang Chen
- Division of Family Medicine
- Division of Geriatric Medicine, Department of Family and Community Medicine, Tri-Service General Hospital, and School of Medicine, National Defense Medical Center, Taipei, Taiwan, Republic of China
| |
Collapse
|
48
|
Alkozi HA, Navarro G, Franco R, Pintor J. Melatonin and the control of intraocular pressure. Prog Retin Eye Res 2019; 75:100798. [PMID: 31560946 DOI: 10.1016/j.preteyeres.2019.100798] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 09/13/2019] [Accepted: 09/18/2019] [Indexed: 12/15/2022]
Abstract
Melatonin is not only synthesized by the pineal gland but by several ocular structures. This natural indoleamine is of great importance for regulating several eye processes, among which pressure homeostasis is included. Glaucoma, the most prevalent eye disease, also known as the silent thief of vision, is a multifactorial pathology that is associated to age and, often, to intraocular hypertension (IOP). Indeed IOP is the only modifiable risk factor and as such medications are available to control it; however, novel medications are sought to minimize undesirable side effects. Melatonin and analogues decrease IOP in both normotensive and hypertensive eyes. Melatonin activates its cognate membrane receptors, MT1 and MT2, which are present in numerous ocular tissues, including the aqueous-humor-producing ciliary processes. Melatonin receptors belong to the superfamily of G-protein-coupled receptors and their activation would lead to different signalling pathways depending on the tissue. This review describes the molecular mechanisms underlying differential functionalities that are attributed to melatonin receptors. Accordingly, the current work highlights the important role of melatonin and its analogues in the healthy and in the glaucomatous eyes, with special attention to the control of intraocular pressure.
Collapse
Affiliation(s)
- Hanan Awad Alkozi
- Department of Biochemistry and Molecular Biology, Faculty of Optics and Optometry, University Complutense of Madrid, Madrid, Spain
| | - Gemma Navarro
- Centro de Investigación en Red, Enfermedades Neurodegeneratives (CiberNed), Instituto de Salud Carlos III, Sinesio Delgado 6, 28029, Madrid, Spain; Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Universitat de Barcelona, Avda. Juan XXIII, 27, 08027, Barcelona, Spain
| | - Rafael Franco
- Centro de Investigación en Red, Enfermedades Neurodegeneratives (CiberNed), Instituto de Salud Carlos III, Sinesio Delgado 6, 28029, Madrid, Spain; Department of Biochemistry and Molecular Biomedicine, School of Biology, Universitat de Barcelona, Diagonal 643, 08028, Barcelona, Barcelona, Spain.
| | - Jesus Pintor
- Department of Biochemistry and Molecular Biology, Faculty of Optics and Optometry, University Complutense of Madrid, Madrid, Spain; Real Academia Nacional de Farmacia, Calle Farmacia 11, 28004, Madrid, Spain.
| |
Collapse
|
49
|
King BJ, Burns SA, Sapoznik KA, Luo T, Gast TJ. High-Resolution, Adaptive Optics Imaging of the Human Trabecular Meshwork In Vivo. Transl Vis Sci Technol 2019; 8:5. [PMID: 31588370 PMCID: PMC6753965 DOI: 10.1167/tvst.8.5.5] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 06/24/2019] [Indexed: 12/22/2022] Open
Abstract
Purpose To image the human trabecular meshwork (TM) in vivo using adaptive optics gonioscopy (AOG) with approximately 2-μm lateral resolution. Methods An existing Indiana University adaptive optics scanning laser ophthalmoscope was altered by adding a 12-mm button lens to a clinical gonioscopic lens allowing high-resolution imaging of the human iridocorneal angle. First an anatomic model eye was used to refine the imaging technique and then nine participants (7 controls and 2 participants with pigment dispersion syndrome) were imaged. Results All nine participants were successfully imaged without adverse events. High-resolution imaging of the human TM was achieved allowing for visualization of the TM beams, and presumed endothelial cells. Uveal meshwork beams in controls averaged 25.5 μm (range, 15.2–44.7) in diameter with pores averaging 42.6 μm (range, 22.3–51.4) while the corneoscleral meshwork pores averaged 8.9 μm (range, 7.7–12.1). Differences in appearance of the uveal and corneoscleral meshwork were noted between the two participants with pigment dispersion syndrome and the controls. These included nearly absent spacing between the beams and enlarged endothelial cells with hyperreflective areas. Conclusions AOG allows for near cellular level resolution of the human TM in vivo. This may allow for further understanding of age-related changes that occur as well as provide a deeper understanding of medical and surgical alterations for the treatment of glaucoma. Translational Relevance Further development of this approach may allow for direct measurements at a micometer level in vivo of changes that occur in the human trabecular meshwork with glaucoma and therapeutic interventions.
Collapse
Affiliation(s)
- Brett J King
- School of Optometry, Indiana University, Bloomington, IN, USA
| | - Stephen A Burns
- School of Optometry, Indiana University, Bloomington, IN, USA
| | | | - Ting Luo
- School of Optometry, Indiana University, Bloomington, IN, USA
| | - Thomas J Gast
- School of Optometry, Indiana University, Bloomington, IN, USA
| |
Collapse
|
50
|
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
|