1
|
Hill KR, Scelsi HF, Youngblood HA, Faralli JA, Itakura T, Fini ME, Peters DM, Lieberman RL. Structural basis for anomalous cellular trafficking behavior of glaucoma-associated A427T mutant myocilin. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.26.640437. [PMID: 40060664 PMCID: PMC11888440 DOI: 10.1101/2025.02.26.640437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/15/2025]
Abstract
Familial mutations in myocilin cause vision loss in glaucoma due to misfolding and a toxic gain of function in a senescent cell type in the anterior eye. Here we characterize the cellular behavior and structure of the myocilin (myocilin A427T) mutant, of uncertain pathogenicity. Our characterization of A427T demonstrates that even mutations that minimally perturb myocilin structure and stability can present challenges for protein quality control clearance pathways. Namely, when expressed in an inducible immortalized trabecular meshwork cell line, inhibition of the proteasome reroutes wild-type myocilin, but not myocilin A427T, from endoplasmic reticulum associated degradation to lysosomal degradation. Yet, the crystal structure of the A427T myocilin olfactomedin domain shows modest perturbations largely confined to the mutation site. The previously unappreciated range of mutant myocilin behavior correlating with variable stability and structure provides a rationale for why it is challenging to predict causal pathogenicity of a given myocilin mutation, even in the presence of clinical data for members of an affected family. Comprehending the continuum of mutant myocilin behavior in the laboratory supports emerging efforts to use genetics to assess glaucoma risk in the clinic. In addition, the study supports a therapeutic strategy aimed at enhancing autophagic clearance of mutant myocilin.
Collapse
Affiliation(s)
- Kamisha R Hill
- School of Chemistry & Biochemistry, Georgia Institute of Technology, Atlanta, GA
| | - Hailee F Scelsi
- School of Chemistry & Biochemistry, Georgia Institute of Technology, Atlanta, GA
| | - Hannah A Youngblood
- School of Chemistry & Biochemistry, Georgia Institute of Technology, Atlanta, GA
| | - Jennifer A Faralli
- Department of Pathology & Laboratory Medicine, University of Wisconsin, Madison, WI
| | - Tatsuo Itakura
- USC Institute for Genetic Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA
| | - M Elizabeth Fini
- New England Eye Center, Tufts Medical Center; Department of Ophthalmology, School of Medicine and Tufts Graduate School of Biomedical Sciences, Tufts University, Boston, MA
| | - Donna M Peters
- Department of Pathology & Laboratory Medicine, University of Wisconsin, Madison, WI
| | - Raquel L Lieberman
- School of Chemistry & Biochemistry, Georgia Institute of Technology, Atlanta, GA
| |
Collapse
|
2
|
Watanabe M, Sato T, Yano T, Higashide M, Ogawa T, Nishikiori N, Furuhashi M, Ohguro H. mTOR Inhibitors Modulate the Biological Nature of TGF-β2-Treated or -Untreated Human Trabecular Meshwork Cells in Different Manners. Biomedicines 2024; 12:2604. [PMID: 39595170 PMCID: PMC11591778 DOI: 10.3390/biomedicines12112604] [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: 10/10/2024] [Revised: 11/05/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
Background/Objectives: Mammalian target of rapamycin (mTOR) inhibition may have been suggested to have a beneficial effect on the glaucomatous human trabecular meshwork (HTM). To study the effects of the mTOR inhibitors rapamycin (Rapa) and Torin1 on the glaucomatous HTM, transforming growth factor-β2 (TGF-β2)-treated two-dimensionally (2D) and three-dimensionally (3D) cultured HTM cells were used. Methods: We evaluated (1) the levels of autophagy via Western blot analysis using a specific antibody against microtubule-associated protein 1 light chain 3 (LC3), (2) barrier capacity based on transepithelial electrical resistance (TEER) and fluorescein isothiocyanate (FITC) permeability (2D), (3) cellular metabolic functions (2D), (4) the size and stiffness of spheroids, and (5) the mRNA expression of ECM proteins. Results: TGF-β2-induced inhibition of autophagy was significantly inhibited by Rapa and Torin1. Rapa and Torin1 substantially decreased barrier capacity in both TGF-β2-untreated and TGF-β2-treated HTM cells. Cellular metabolic analysis indicated that Rapa, but not Torin1, substantially enhanced both mitochondrial and glycolytic functions of TGF-β2-untreated HTM cells. In the physical properties of spheroids, TGF-β2 resulted in the formation of down-sized and stiffened spheroids. mTOR inhibitors decreased the size but not the stiffness of TGF-β2-untreated spheroids and significantly reduced the TGF-β2-related increase in the stiffness but not the size of spheroids. The diverse effects of mTOR inhibitors on TGF-β2-untreated and TGF-β2-treated spheroids were also observed in the mRNA expression of extracellular matrix proteins. Conclusions: The results taken together suggest that mTOR inhibitors significantly influence the biological aspects of both a single layer and multiple layers of the TGF-β2-treated HTM and untreated HTM.
Collapse
Affiliation(s)
- Megumi Watanabe
- Departments of Ophthalmology, School of Medicine, Sapporo Medical University, S1W17, Sapporo 060-8556, Japan; (M.W.); (M.H.); (N.N.)
| | - Tatsuya Sato
- Departments of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University, S1W17, Sapporo 060-8556, Japan; (T.S.); (T.Y.); (T.O.); (M.F.)
- Departments of Cellular Physiology and Signal Transduction, Sapporo Medical University, S1W17, Sapporo 060-8556, Japan
| | - Toshiyuki Yano
- Departments of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University, S1W17, Sapporo 060-8556, Japan; (T.S.); (T.Y.); (T.O.); (M.F.)
| | - Megumi Higashide
- Departments of Ophthalmology, School of Medicine, Sapporo Medical University, S1W17, Sapporo 060-8556, Japan; (M.W.); (M.H.); (N.N.)
| | - Toshifumi Ogawa
- Departments of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University, S1W17, Sapporo 060-8556, Japan; (T.S.); (T.Y.); (T.O.); (M.F.)
- Departments of Cellular Physiology and Signal Transduction, Sapporo Medical University, S1W17, Sapporo 060-8556, Japan
| | - Nami Nishikiori
- Departments of Ophthalmology, School of Medicine, Sapporo Medical University, S1W17, Sapporo 060-8556, Japan; (M.W.); (M.H.); (N.N.)
| | - Masato Furuhashi
- Departments of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University, S1W17, Sapporo 060-8556, Japan; (T.S.); (T.Y.); (T.O.); (M.F.)
| | - Hiroshi Ohguro
- Departments of Ophthalmology, School of Medicine, Sapporo Medical University, S1W17, Sapporo 060-8556, Japan; (M.W.); (M.H.); (N.N.)
| |
Collapse
|
3
|
Tundo GR, Cavaterra D, Pandino I, Zingale GA, Giammaria S, Boccaccini A, Michelessi M, Roberti G, Tanga L, Carnevale C, Figus M, Grasso G, Coletta M, Bocedi A, Oddone F, Sbardella D. The Delayed Turnover of Proteasome Processing of Myocilin upon Dexamethasone Stimulation Introduces the Profiling of Trabecular Meshwork Cells' Ubiquitylome. Int J Mol Sci 2024; 25:10017. [PMID: 39337505 PMCID: PMC11432723 DOI: 10.3390/ijms251810017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/11/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
Glaucoma is chronic optic neuropathy whose pathogenesis has been associated with the altered metabolism of Trabecular Meshwork Cells, which is a cell type involved in the synthesis and remodeling of the trabecular meshwork, the main drainage pathway of the aqueous humor. Starting from previous findings supporting altered ubiquitin signaling, in this study, we investigated the ubiquitin-mediated turnover of myocilin (MYOC/TIGR gene), which is a glycoprotein with a recognized role in glaucoma pathogenesis, in a human Trabecular Meshwork strain cultivated in vitro in the presence of dexamethasone. This is a validated experimental model of steroid-induced glaucoma, and myocilin upregulation by glucocorticoids is a phenotypic marker of Trabecular Meshwork strains. Western blotting and native-gel electrophoresis first uncovered that, in the presence of dexamethasone, myocilin turnover by proteasome particles was slower than in the absence of the drug. Thereafter, co-immunoprecipitation, RT-PCR and gene-silencing studies identified STUB1/CHIP as a candidate E3-ligase of myocilin. In this regard, dexamethasone treatment was found to downregulate STUB1/CHIP levels by likely promoting its proteasome-mediated turnover. Hence, to strengthen the working hypothesis about global alterations of ubiquitin-signaling, the first profiling of TMCs ubiquitylome, in the presence and absence of dexamethasone, was here undertaken by diGLY proteomics. Application of this workflow effectively highlighted a robust dysregulation of key pathways (e.g., phospholipid signaling, β-catenin, cell cycle regulation) in dexamethasone-treated Trabecular Meshwork Cells, providing an ubiquitin-centered perspective around the effect of glucocorticoids on metabolism and glaucoma pathogenesis.
Collapse
Affiliation(s)
- Grazia Raffaella Tundo
- Department of Clinical Sciences and Translational Medicine, University of Tor Vergata, 00133 Rome, Italy
| | - Dario Cavaterra
- Department of Chemical Sciences and Technologies, University of Tor Vergata, 00133 Rome, Italy (A.B.)
| | - Irene Pandino
- IRCCS-Fondazione Bietti, 00168 Rome, Italy (G.R.); (M.C.)
| | | | - Sara Giammaria
- IRCCS-Fondazione Bietti, 00168 Rome, Italy (G.R.); (M.C.)
| | | | | | - Gloria Roberti
- IRCCS-Fondazione Bietti, 00168 Rome, Italy (G.R.); (M.C.)
| | - Lucia Tanga
- IRCCS-Fondazione Bietti, 00168 Rome, Italy (G.R.); (M.C.)
| | | | - Michele Figus
- Department of Surgical, Medical, Molecular Pathology and Critical Care Medicine, University of Pisa, 56124 Pisa, Italy;
| | - Giuseppe Grasso
- Department of Chemical Sciences, University of Catania, 95125 Catania, Italy;
| | | | - Alessio Bocedi
- Department of Chemical Sciences and Technologies, University of Tor Vergata, 00133 Rome, Italy (A.B.)
| | | | | |
Collapse
|
4
|
Becker S, L'Ecuyer Z, Jones BW, Zouache MA, McDonnell FS, Vinberg F. Modeling complex age-related eye disease. Prog Retin Eye Res 2024; 100:101247. [PMID: 38365085 PMCID: PMC11268458 DOI: 10.1016/j.preteyeres.2024.101247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 01/31/2024] [Accepted: 02/02/2024] [Indexed: 02/18/2024]
Abstract
Modeling complex eye diseases like age-related macular degeneration (AMD) and glaucoma poses significant challenges, since these conditions depend highly on age-related changes that occur over several decades, with many contributing factors remaining unknown. Although both diseases exhibit a relatively high heritability of >50%, a large proportion of individuals carrying AMD- or glaucoma-associated genetic risk variants will never develop these diseases. Furthermore, several environmental and lifestyle factors contribute to and modulate the pathogenesis and progression of AMD and glaucoma. Several strategies replicate the impact of genetic risk variants, pathobiological pathways and environmental and lifestyle factors in AMD and glaucoma in mice and other species. In this review we will primarily discuss the most commonly available mouse models, which have and will likely continue to improve our understanding of the pathobiology of age-related eye diseases. Uncertainties persist whether small animal models can truly recapitulate disease progression and vision loss in patients, raising doubts regarding their usefulness when testing novel gene or drug therapies. We will elaborate on concerns that relate to shorter lifespan, body size and allometries, lack of macula and a true lamina cribrosa, as well as absence and sequence disparities of certain genes and differences in their chromosomal location in mice. Since biological, rather than chronological, age likely predisposes an organism for both glaucoma and AMD, more rapidly aging organisms like small rodents may open up possibilities that will make research of these diseases more timely and financially feasible. On the other hand, due to the above-mentioned anatomical and physiological features, as well as pharmacokinetic and -dynamic differences small animal models are not ideal to study the natural progression of vision loss or the efficacy and safety of novel therapies. In this context, we will also discuss the advantages and pitfalls of alternative models that include larger species, such as non-human primates and rabbits, patient-derived retinal organoids, and human organ donor eyes.
Collapse
Affiliation(s)
- Silke Becker
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA
| | - Zia L'Ecuyer
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA
| | - Bryan W Jones
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA
| | - Moussa A Zouache
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA
| | - Fiona S McDonnell
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA; Biomedical Engineering, University of Utah, Salt Lake City, UT, USA
| | - Frans Vinberg
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA; Biomedical Engineering, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
5
|
Yan X, Wu S, Liu Q, Cheng Y, Teng Y, Ren T, Zhang J, Wang N. Serine to proline mutation at position 341 of MYOC impairs trabecular meshwork function by causing autophagy deregulation. Cell Death Discov 2024; 10:21. [PMID: 38212635 PMCID: PMC10784477 DOI: 10.1038/s41420-024-01801-1] [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: 06/30/2023] [Revised: 12/24/2023] [Accepted: 01/04/2024] [Indexed: 01/13/2024] Open
Abstract
Glaucoma is a highly heritable disease, and myocilin was the first identified causal and most common pathogenic gene in glaucoma. Serine-to-proline mutation at position 341 of myocilin (MYOCS341P) is associated with severe glaucoma phenotypes in a five-generation primary open-angle glaucoma family. However, the underlying mechanisms are underexplored. Herein, we established the MYOCS341P transgenic mouse model and characterized the glaucoma phenotypes. Further, we systematically explored the functional differences between wild-type and MYOCS341P through immunoprecipitation, mass spectrometry, and RNA-seq analyses. We found that MYOCS341P transgenic mice exhibit glaucoma phenotypes, characterized by reduced aqueous humor outflow, elevated intraocular pressure, decreased trabecular meshwork (TM) cell number, narrowed Schlemm's canal, retinal ganglion cell loss, and visual impairment. Mechanistically, the secretion of dysfunctional MYOCS341P accumulated in the endoplasmic reticulum (ER), inducing ER stress and dysregulation of autophagy, thereby promoting TM cell death. We describe an effective transgenic model for mechanistic studies and the screening of therapeutic targets. Our data generated from high-throughput analyses help elucidate the mechanism underlying mutant MYOC-related glaucoma.
Collapse
Affiliation(s)
- Xuejing Yan
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University; Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing, 100730, China
- Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, 100069, China
| | - Shen Wu
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University; Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing, 100730, China
- Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, 100069, China
| | - Qian Liu
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University; Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing, 100730, China
- Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, 100069, China
| | - Ying Cheng
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University; Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing, 100730, China
| | - Yufei Teng
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University; Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing, 100730, China
| | - Tianmin Ren
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University; Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing, 100730, China
| | - Jingxue Zhang
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University; Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing, 100730, China.
- Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, 100069, China.
| | - Ningli Wang
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University; Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing, 100730, China.
- Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
6
|
Saccuzzo EG, Mebrat MD, Scelsi HF, Kim M, Ma MT, Su X, Hill SE, Rheaume E, Li R, Torres MP, Gumbart JC, Van Horn WD, Lieberman RL. Competition between inside-out unfolding and pathogenic aggregation in an amyloid-forming β-propeller. Nat Commun 2024; 15:155. [PMID: 38168102 PMCID: PMC10762032 DOI: 10.1038/s41467-023-44479-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 12/14/2023] [Indexed: 01/05/2024] Open
Abstract
Studies of folded-to-misfolded transitions using model protein systems reveal a range of unfolding needed for exposure of amyloid-prone regions for subsequent fibrillization. Here, we probe the relationship between unfolding and aggregation for glaucoma-associated myocilin. Mutations within the olfactomedin domain of myocilin (OLF) cause a gain-of-function, namely cytotoxic intracellular aggregation, which hastens disease progression. Aggregation by wild-type OLF (OLFWT) competes with its chemical unfolding, but only below the threshold where OLF loses tertiary structure. Representative moderate (OLFD380A) and severe (OLFI499F) disease variants aggregate differently, with rates comparable to OLFWT in initial stages of unfolding, and variants adopt distinct partially folded structures seen along the OLFWT urea-unfolding pathway. Whether initiated with mutation or chemical perturbation, unfolding propagates outward to the propeller surface. In sum, for this large protein prone to amyloid formation, the requirement for a conformational change to promote amyloid fibrillization leads to direct competition between unfolding and aggregation.
Collapse
Affiliation(s)
- Emily G Saccuzzo
- School of Chemistry & Biochemistry, Georgia Institute of Technology, Atlanta, USA
| | - Mubark D Mebrat
- Biodesign Center for Personalized Diagnostics, Arizona State University, Tempe, USA
- School of Molecular Sciences, Arizona State University, Tempe, USA
| | - Hailee F Scelsi
- School of Chemistry & Biochemistry, Georgia Institute of Technology, Atlanta, USA
| | - Minjoo Kim
- Biodesign Center for Personalized Diagnostics, Arizona State University, Tempe, USA
- School of Molecular Sciences, Arizona State University, Tempe, USA
| | - Minh Thu Ma
- School of Chemistry & Biochemistry, Georgia Institute of Technology, Atlanta, USA
| | - Xinya Su
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, USA
| | - Shannon E Hill
- School of Chemistry & Biochemistry, Georgia Institute of Technology, Atlanta, USA
| | - Elisa Rheaume
- Interdisciplinary Graduate Program in Quantitative Biosciences, Georgia Institute of Technology, Atlanta, USA
| | - Renhao Li
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta and Department of Pediatrics, Emory University School of Medicine, Atlanta, USA
| | - Matthew P Torres
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, USA
| | - James C Gumbart
- School of Chemistry & Biochemistry, Georgia Institute of Technology, Atlanta, USA
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, USA
- School of Physics, Georgia Institute of Technology, Atlanta, USA
| | - Wade D Van Horn
- Biodesign Center for Personalized Diagnostics, Arizona State University, Tempe, USA.
- School of Molecular Sciences, Arizona State University, Tempe, USA.
| | - Raquel L Lieberman
- School of Chemistry & Biochemistry, Georgia Institute of Technology, Atlanta, USA.
| |
Collapse
|
7
|
Shim MS, Liton PB. The physiological and pathophysiological roles of the autophagy lysosomal system in the conventional aqueous humor outflow pathway: More than cellular clean up. Prog Retin Eye Res 2022; 90:101064. [PMID: 35370083 PMCID: PMC9464695 DOI: 10.1016/j.preteyeres.2022.101064] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 03/09/2022] [Accepted: 03/25/2022] [Indexed: 10/18/2022]
Abstract
During the last few years, the autophagy lysosomal system is emerging as a central cellular pathway with roles in survival, acting as a housekeeper and stress response mechanism. Studies by our and other labs suggest that autophagy might play an essential role in maintaining aqueous humor outflow homeostasis, and that malfunction of autophagy in outflow pathway cells might predispose to ocular hypertension and glaucoma pathogenesis. In this review, we will collect the current knowledge and discuss the molecular mechanisms by which autophagy does or might regulate normal outflow pathway tissue function, and its response to different types of stressors (oxidative stress and mechanical stress). We will also discuss novel roles of autophagy and lysosomal enzymes in modulation of TGFβ signaling and ECM remodeling, and the link between dysregulated autophagy and cellular senescence. We will examine what we have learnt, using pre-clinical animal models about how dysregulated autophagy can contribute to disease and apply that to the current status of autophagy in human glaucoma. Finally, we will consider and discuss the challenges and the potential of autophagy as a therapeutic target for the treatment of ocular hypertension and glaucoma.
Collapse
Affiliation(s)
- Myoung Sup Shim
- Duke University, Department of Ophthalmology, Durham, NC, 27705, USA
| | - Paloma B Liton
- Duke University, Department of Ophthalmology, Durham, NC, 27705, USA.
| |
Collapse
|
8
|
Yan X, Wu S, Liu Q, Cheng Y, Zhang J, Wang N. Myocilin Gene Mutation Induced Autophagy Activation Causes Dysfunction of Trabecular Meshwork Cells. Front Cell Dev Biol 2022; 10:900777. [PMID: 35615698 PMCID: PMC9124892 DOI: 10.3389/fcell.2022.900777] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 04/08/2022] [Indexed: 12/12/2022] Open
Abstract
Trabecular meshwork dysfunction is the main cause of primary open angle glaucoma (POAG) associated with elevated intraocular pressure (IOP). Mutant myocilin causes glaucoma mainly via elevating IOP. Previously we have found that accumulated Asn 450 Tyr (N450Y) mutant myocilin impairs human trabecular meshwork (TM) cells by inducing chronic endoplasmic reticulum (ER) stress response in vitro. However, it is unclear how ER stress leads to TM damage and whether N450Y myocilin mutation is associated with POAG in vivo. Here we found that N450Y mutant myocilin induces autophagy, which worsens cell viability, whereas inhibition of autophagy increases viability and decreases cell death in human TM cells. Furthermore, we construct a transgenic mouse model of N450Y myocilin mutation (Tg-MYOCN450Y) and Tg-MYOCN450Y mice exhibiting glaucoma phenotypes: IOP elevation, retinal ganglion cell loss and visual impairment. Consistent with our published in vitro studies, mutant myocilin fails to secrete into aqueous humor, causes ER stress and actives autophagy in Tg-MYOCN450Y mice, and aqueous humor dynamics are altered in Tg-MYOCN450Y mice. In summary, our studies demonstrate that activation of autophagy is correlated with pathogenesis of POAG.
Collapse
Affiliation(s)
- Xuejing Yan
- Beijing Tongren Eye Center, Beijing Ophthalmology and Visual Sciences Key Laboratory, Beijing Tongren Hospital, Beijing Institute of Ophthalmology, Capital Medical University, Beijing, China
- Collaborative Innovation Center for Brain Disorders, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Shen Wu
- Beijing Tongren Eye Center, Beijing Ophthalmology and Visual Sciences Key Laboratory, Beijing Tongren Hospital, Beijing Institute of Ophthalmology, Capital Medical University, Beijing, China
- Collaborative Innovation Center for Brain Disorders, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Qian Liu
- Beijing Tongren Eye Center, Beijing Ophthalmology and Visual Sciences Key Laboratory, Beijing Tongren Hospital, Beijing Institute of Ophthalmology, Capital Medical University, Beijing, China
- Collaborative Innovation Center for Brain Disorders, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Ying Cheng
- Beijing Tongren Eye Center, Beijing Ophthalmology and Visual Sciences Key Laboratory, Beijing Tongren Hospital, Beijing Institute of Ophthalmology, Capital Medical University, Beijing, China
| | - Jingxue Zhang
- Beijing Tongren Eye Center, Beijing Ophthalmology and Visual Sciences Key Laboratory, Beijing Tongren Hospital, Beijing Institute of Ophthalmology, Capital Medical University, Beijing, China
- Collaborative Innovation Center for Brain Disorders, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
- *Correspondence: Ningli Wang, ; Jingxue Zhang,
| | - Ningli Wang
- Beijing Tongren Eye Center, Beijing Ophthalmology and Visual Sciences Key Laboratory, Beijing Tongren Hospital, Beijing Institute of Ophthalmology, Capital Medical University, Beijing, China
- Collaborative Innovation Center for Brain Disorders, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
- *Correspondence: Ningli Wang, ; Jingxue Zhang,
| |
Collapse
|
9
|
Liang X, Li N, Rong Y, Wang J, Zhang H. Identification of proteomic changes for dexamethasone-induced ocular hypertension using a tandem mass tag (TMT) approach. Exp Eye Res 2021; 216:108914. [PMID: 34979099 DOI: 10.1016/j.exer.2021.108914] [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: 05/11/2021] [Revised: 12/10/2021] [Accepted: 12/23/2021] [Indexed: 11/29/2022]
Abstract
Glaucoma, characterized by ocular hypertension, is the second most common cause of vision loss worldwide. The potential mechanism, however, has yet to be elucidated. This study aimed to assess the proteomic changes in the trabecular meshwork (TM) in an observational animal model of Dexamethasone (DEX)-induced OHT. OHT was induced in Wistar rats by applying DEX topically to both eyes for 28 days. Intraocular pressure (IOP) was evaluated and TM protein expressions and protein identification were performed by a TMT-based method for comparing the changes in proteins between DEX-induced OHT and the control group. The results showed that average IOP was elevated significantly in rats of the DEX-induced OHT group compared to controls. Further, a total of 4,804 proteins in the control and DEX-induced OHT group were determined and 4,064 proteins were quantified via TMT proteomics. In total, 292 significantly abundant proteins (173 downregulated and 119 upregulated) were identified between the two groups. Proteins associated with vision, including Crystallin related proteins, filensin, rhodopsin, recoverin, phosducin were lowered in the DEX-induced OHT group relative to the control group. In summary, DEX induced extensive changes in the protein expression of TM tissue. These proteins were found to be candidate biomarkers for personalized treatment and diagnostic research in the future for improving visual health.
Collapse
Affiliation(s)
- Xin Liang
- Department of Ophthalmology, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ning Li
- Department of Ophthalmology, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yan Rong
- Department of Ophthalmology, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Junming Wang
- Department of Ophthalmology, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei, China.
| | - Hong Zhang
- Department of Ophthalmology, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
10
|
Sharma R, Grover A. Myocilin-associated Glaucoma: A Historical Perspective and Recent Research Progress. Mol Vis 2021; 27:480-493. [PMID: 34497454 PMCID: PMC8403517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 08/18/2021] [Indexed: 10/29/2022] Open
Abstract
Glaucoma a debilitating disease, is globally the second most common kind of permanent blindness. Primary open-angle glaucoma (POAG) is its most prevalent form and is often linked with alterations in the myocilin gene (MYOC). MYOC encodes the myocilin protein, which is expressed throughout the body, but primarily in trabecular meshwork (TM) tissue in the eyes. TM is principally involved in regulating intraocular pressure (IOP), and elevated IOP is the main risk factor associated with glaucoma. The myocilin protein's function remains unknown; however, mutations compromise its folding and processing inside TM cells, contributing to the glaucoma phenotype. While glaucoma is a complex disease with various molecules and factors as contributing causes, the role played by myocilin has been the most widely studied. The current review describes the present understanding of myocilin and its association with glaucoma and aims to shift the focus toward developing targeted therapies for treating glaucoma patients with variations in MYOC.
Collapse
|
11
|
Sbardella D, Tundo GR, Coletta M, Manni G, Oddone F. Dexamethasone Downregulates Autophagy through Accelerated Turn-Over of the Ulk-1 Complex in a Trabecular Meshwork Cells Strain: Insights on Steroid-Induced Glaucoma Pathogenesis. Int J Mol Sci 2021; 22:ijms22115891. [PMID: 34072647 PMCID: PMC8198647 DOI: 10.3390/ijms22115891] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 05/20/2021] [Accepted: 05/24/2021] [Indexed: 12/16/2022] Open
Abstract
Steroid-induced glaucoma is a severe pathological condition, sustained by a rapidly progressive increase in intraocular pressure (IOP), which is diagnosed in a subset of subjects who adhere to a glucocorticoid (GC)-based therapy. Molecular and clinical studies suggest that either natural or synthetic GCs induce a severe metabolic dysregulation of Trabecular Meshwork Cells (TMCs), an endothelial-derived histotype with phagocytic and secretive functions which lay at the iridocorneal angle in the anterior segment of the eye. Since TMCs physiologically regulate the composition and architecture of trabecular meshwork (TM), which is the main outflow pathway of aqueous humor, a fluid which shapes the eye globe and nourishes the lining cell types, GCs are supposed to trigger a pathological remodeling of the TM, inducing an IOP increase and retina mechanical compression. The metabolic dysregulation of TMCs induced by GCs exposure has never been characterized at the molecular detail. Herein, we report that, upon dexamethasone exposure, a TMCs strain develops a marked inhibition of the autophagosome biogenesis pathway through an enhanced turnover of two members of the Ulk-1 complex, the main platform for autophagy induction, through the Ubiquitin Proteasome System (UPS).
Collapse
Affiliation(s)
- Diego Sbardella
- IRCCS-Fondazione Bietti, 00198 Rome, Italy;
- Correspondence: (D.S.); (F.O.)
| | | | - Massimo Coletta
- Department of Clinical Sciences and Translational Medicine, University of Tor Vergata, 00133 Rome, Italy; (M.C.); (G.M.)
| | - Gianluca Manni
- Department of Clinical Sciences and Translational Medicine, University of Tor Vergata, 00133 Rome, Italy; (M.C.); (G.M.)
| | - Francesco Oddone
- IRCCS-Fondazione Bietti, 00198 Rome, Italy;
- Correspondence: (D.S.); (F.O.)
| |
Collapse
|
12
|
Kasetti RB, Maddineni P, Kiehlbauch C, Patil S, Searby CC, Levine B, Sheffield VC, Zode GS. Autophagy stimulation reduces ocular hypertension in a murine glaucoma model via autophagic degradation of mutant myocilin. JCI Insight 2021; 6:143359. [PMID: 33539326 PMCID: PMC8021112 DOI: 10.1172/jci.insight.143359] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 01/28/2021] [Indexed: 02/06/2023] Open
Abstract
Elevation of intraocular pressure (IOP) due to trabecular meshwork (TM) damage is associated with primary open-angle glaucoma (POAG). Myocilin mutations resulting in elevated IOP are the most common genetic causes of POAG. We have previously shown that mutant myocilin accumulates in the ER and induces chronic ER stress, leading to TM damage and IOP elevation. However, it is not understood how chronic ER stress leads to TM dysfunction and loss. Here, we report that mutant myocilin activated autophagy but was functionally impaired in cultured human TM cells and in a mouse model of myocilin-associated POAG (Tg-MYOCY437H). Genetic and pharmacological inhibition of autophagy worsened mutant myocilin accumulation and exacerbated IOP elevation in Tg-MYOCY437H mice. Remarkably, impaired autophagy was associated with chronic ER stress-induced transcriptional factor CHOP. Deletion of CHOP corrected impaired autophagy, enhanced recognition and degradation of mutant myocilin by autophagy, and reduced glaucoma in Tg-MYOCY437H mice. Stimulating autophagic flux via tat-beclin 1 peptide or torin 2 promoted autophagic degradation of mutant myocilin and reduced elevated IOP in Tg-MYOCY437H mice. Our study provides an alternate treatment strategy for myocilin-associated POAG by correcting impaired autophagy in the TM.
Collapse
Affiliation(s)
- Ramesh B. Kasetti
- Department of Pharmacology and Neuroscience and the North Texas Eye Research Institute, University of North Texas Health Science Center at Fort Worth, Fort Worth, Texas, USA
| | - Prabhavathi Maddineni
- Department of Pharmacology and Neuroscience and the North Texas Eye Research Institute, University of North Texas Health Science Center at Fort Worth, Fort Worth, Texas, USA
| | - Charles Kiehlbauch
- Department of Pharmacology and Neuroscience and the North Texas Eye Research Institute, University of North Texas Health Science Center at Fort Worth, Fort Worth, Texas, USA
| | - Shruti Patil
- Department of Pharmacology and Neuroscience and the North Texas Eye Research Institute, University of North Texas Health Science Center at Fort Worth, Fort Worth, Texas, USA
| | - Charles C. Searby
- Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Beth Levine
- Center for Autophagy Research, Department of Internal Medicine
- Howard Hughes Medical Institute, and
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Val C. Sheffield
- Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Gulab S. Zode
- Department of Pharmacology and Neuroscience and the North Texas Eye Research Institute, University of North Texas Health Science Center at Fort Worth, Fort Worth, Texas, USA
| |
Collapse
|
13
|
Tundo GR, Sbardella D, Santoro AM, Coletta A, Oddone F, Grasso G, Milardi D, Lacal PM, Marini S, Purrello R, Graziani G, Coletta M. The proteasome as a druggable target with multiple therapeutic potentialities: Cutting and non-cutting edges. Pharmacol Ther 2020; 213:107579. [PMID: 32442437 PMCID: PMC7236745 DOI: 10.1016/j.pharmthera.2020.107579] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 05/05/2020] [Indexed: 01/10/2023]
Abstract
Ubiquitin Proteasome System (UPS) is an adaptable and finely tuned system that sustains proteostasis network under a large variety of physiopathological conditions. Its dysregulation is often associated with the onset and progression of human diseases; hence, UPS modulation has emerged as a promising new avenue for the development of treatments of several relevant pathologies, such as cancer and neurodegeneration. The clinical interest in proteasome inhibition has considerably increased after the FDA approval in 2003 of bortezomib for relapsed/refractory multiple myeloma, which is now used in the front-line setting. Thereafter, two other proteasome inhibitors (carfilzomib and ixazomib), designed to overcome resistance to bortezomib, have been approved for treatment-experienced patients, and a variety of novel inhibitors are currently under preclinical and clinical investigation not only for haematological malignancies but also for solid tumours. However, since UPS collapse leads to toxic misfolded proteins accumulation, proteasome is attracting even more interest as a target for the care of neurodegenerative diseases, which are sustained by UPS impairment. Thus, conceptually, proteasome activation represents an innovative and largely unexplored target for drug development. According to a multidisciplinary approach, spanning from chemistry, biochemistry, molecular biology to pharmacology, this review will summarize the most recent available literature regarding different aspects of proteasome biology, focusing on structure, function and regulation of proteasome in physiological and pathological processes, mostly cancer and neurodegenerative diseases, connecting biochemical features and clinical studies of proteasome targeting drugs.
Collapse
Affiliation(s)
- G R Tundo
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy.
| | | | - A M Santoro
- CNR, Institute of Crystallography, Catania, Italy
| | - A Coletta
- Department of Chemistry, University of Aarhus, Aarhus, Denmark
| | - F Oddone
- IRCCS-Fondazione Bietti, Rome, Italy
| | - G Grasso
- Department of Chemical Sciences, University of Catania, Catania, Italy
| | - D Milardi
- CNR, Institute of Crystallography, Catania, Italy
| | - P M Lacal
- Laboratory of Molecular Oncology, IDI-IRCCS, Rome, Italy
| | - S Marini
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy
| | - R Purrello
- Department of Chemical Sciences, University of Catania, Catania, Italy
| | - G Graziani
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.
| | - M Coletta
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy.
| |
Collapse
|
14
|
ER-Phagy: Quality Control and Turnover of Endoplasmic Reticulum. Trends Cell Biol 2020; 30:384-398. [PMID: 32302550 DOI: 10.1016/j.tcb.2020.02.001] [Citation(s) in RCA: 190] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 01/30/2020] [Accepted: 02/03/2020] [Indexed: 12/11/2022]
Abstract
The endoplasmic reticulum (ER) is the largest organelle in cells and has fundamental functions, such as folding, processing, and trafficking of proteins, cellular metabolism, and ion storage. To maintain its function, it is turned over constitutively, and even more actively under certain stress conditions. Quality control of the ER is mediated primarily by two pathways: the ubiquitin-proteasome system and autophagy (termed 'ER-phagy'). The identification of ER-phagy adaptor molecules has shed light on the mechanisms and physiological significance of ER-phagy. Here, we describe recent findings on various types of ER-phagy and present unanswered questions related to their mechanism and regulation.
Collapse
|
15
|
Takahashi Y, Liang X, Hattori T, Tang Z, He H, Chen H, Liu X, Abraham T, Imamura-Kawasawa Y, Buchkovich NJ, Young MM, Wang HG. VPS37A directs ESCRT recruitment for phagophore closure. J Cell Biol 2019; 218:3336-3354. [PMID: 31519728 PMCID: PMC6781443 DOI: 10.1083/jcb.201902170] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 06/25/2019] [Accepted: 07/26/2019] [Indexed: 12/11/2022] Open
Abstract
Takahashi et al. perform a genome-wide CRISPR screen using the HaloTag-LC3 assay to gain insight into the mechanisms of phagophore closure. They identify a role for VPS37A in coordinating the ESCRT assembly on the phagophore for membrane closure. The process of phagophore closure requires the endosomal sorting complex required for transport III (ESCRT-III) subunit CHMP2A and the AAA ATPase VPS4, but their regulatory mechanisms remain unknown. Here, we establish a FACS-based HaloTag-LC3 autophagosome completion assay to screen a genome-wide CRISPR library and identify the ESCRT-I subunit VPS37A as a critical component for phagophore closure. VPS37A localizes on the phagophore through the N-terminal putative ubiquitin E2 variant domain, which is found to be required for autophagosome completion but dispensable for ESCRT-I complex formation and the degradation of epidermal growth factor receptor in the multivesicular body pathway. Notably, loss of VPS37A abrogates the phagophore recruitment of the ESCRT-I subunit VPS28 and CHMP2A, whereas inhibition of membrane closure by CHMP2A depletion or VPS4 inhibition accumulates VPS37A on the phagophore. These observations suggest that VPS37A coordinates the recruitment of a unique set of ESCRT machinery components for phagophore closure in mammalian cells.
Collapse
Affiliation(s)
| | - Xinwen Liang
- Department of Pediatrics, Penn State College of Medicine, Hershey, PA
| | - Tatsuya Hattori
- Department of Pediatrics, Penn State College of Medicine, Hershey, PA
| | - Zhenyuan Tang
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA
| | - Haiyan He
- Department of Pediatrics, Penn State College of Medicine, Hershey, PA
| | - Han Chen
- Microscopy Imaging Facility, Penn State College of Medicine, Hershey, PA
| | - Xiaoming Liu
- Department of Pediatrics, Penn State College of Medicine, Hershey, PA
| | - Thomas Abraham
- Department of Neural and Behavioral Science, Penn State College of Medicine, Hershey, PA.,Microscopy Imaging Facility, Penn State College of Medicine, Hershey, PA
| | - Yuka Imamura-Kawasawa
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA.,Institute for Personalized Medicine, Penn State College of Medicine, Hershey, PA
| | - Nicholas J Buchkovich
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, PA
| | - Megan M Young
- Department of Pediatrics, Penn State College of Medicine, Hershey, PA
| | - Hong-Gang Wang
- Department of Pediatrics, Penn State College of Medicine, Hershey, PA .,Department of Pharmacology, Penn State College of Medicine, Hershey, PA
| |
Collapse
|
16
|
Abstract
Many diseases are related to age, among these neurodegeneration is particularly important. Alzheimer's disease Parkinson's and Glaucoma have many common pathogenic events including oxidative damage, Mitochondrial dysfunction, endothelial alterations and changes in the visual field. These are well known in the case of glaucoma, less in the case of neurodegeneration of the brain. Many other molecular aspects are common, such as the role of endoplasmic reticulum autophagy and neuronal apoptosis while others have been neglected due to lack of space such as inflammatory cytokine or miRNA. Moreover, the loss of specific neuronal populations, the induction of similar mechanisms of cell injury and the deposition of protein aggregates in specific anatomical areas are very similar events between these diseases. Intracellular and/or extracellular accumulation of protein aggregates is a key feature of many neurodegenerative disorders. The existence of abnormal protein aggregates has been documented in the RGCs of glaucomatous patients such as the anomalous Tau protein or the β-amyloid accumulations. Intra-cell catabolic processes also appear to be common in both glaucoma and neurodegeneration. They also help us to understand how the basis between these diseases is common and how the visual aspects can be a serious problem for those who are affected.
Collapse
Affiliation(s)
- Sergio Claudio Saccà
- Department of Head/Neck Pathologies, St Martino Hospital, Ophthalmology Unit, Genoa, Italy.
| | - Carlo Alberto Cutolo
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics and Maternal and Child Science, University of Genoa, Policlinico San Martino Hospital, Eye Clinic Genoa, Genoa, Italy
| | - Tommaso Rossi
- Department of Head/Neck Pathologies, St Martino Hospital, Ophthalmology Unit, Genoa, Italy
| |
Collapse
|
17
|
Wang H, Li M, Zhang Z, Xue H, Chen X, Ji Y. Physiological function of myocilin and its role in the pathogenesis of glaucoma in the trabecular meshwork (Review). Int J Mol Med 2018; 43:671-681. [PMID: 30483726 PMCID: PMC6317685 DOI: 10.3892/ijmm.2018.3992] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Accepted: 11/16/2018] [Indexed: 12/26/2022] Open
Abstract
Myocilin is highly expressed in the trabecular meshwork (TM), which plays an important role in the regulation of intraocular pressure (IOP). Myocilin abnormalities may cause dysfunction of the TM, potentially leading to increased IOP. High IOP is a well‑known primary risk factor for glaucoma. Myocilin mutations are common among glaucoma patients, and they are implicated in juvenile‑onset open‑angle glaucoma (JOAG) and adult‑onset primary open‑angle glaucoma (POAG). Aggregation of aberrant mutant myocilins is closely associated with glaucoma pathogenesis. The aim of the present review was to discuss the recent findings regarding the major physiological functions of myocilin, such as intra‑ and extracellular proteolytic processes. We also aimed to discuss the risk factors associated with myocilin and the development of glaucoma, such as misfolded/mutant myocilin, imbalance of myocilin and extracellular proteins, and instability of mutant myocilin associated with temperature. Finally, we further outlined certain issues that are yet to be resolved, which may represent the basis for future studies on the role of myocilin in glaucoma.
Collapse
Affiliation(s)
- Hongwei Wang
- Department of Ophthalmology, Jingjiang People's Hospital, Jingjiang, Jiangsu 214500, P.R. China
| | - Mingzhe Li
- Department of Ophthalmology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, P.R. China
| | - Zhenzhen Zhang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Haifeng Xue
- Public Health School, Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - Xing Chen
- Department of Science and Education, Jingjiang People's Hospital, Jingjiang, Jiangsu 214500, P.R. China
| | - Yong Ji
- Department of General Surgery, Jingjiang People's Hospital, Jingjiang, Jiangsu 214500, P.R. China
| |
Collapse
|
18
|
Lynch JM, Li B, Katoli P, Xiang C, Leehy B, Rangaswamy N, Saenz-Vash V, Wang YK, Lei H, Nicholson TB, Meredith E, Rice DS, Prasanna G, Chen A. Binding of a glaucoma-associated myocilin variant to the αB-crystallin chaperone impedes protein clearance in trabecular meshwork cells. J Biol Chem 2018; 293:20137-20156. [PMID: 30389787 DOI: 10.1074/jbc.ra118.004325] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 10/19/2018] [Indexed: 01/09/2023] Open
Abstract
Myocilin (MYOC) was discovered more than 20 years ago and is the gene whose mutations are most commonly observed in individuals with glaucoma. Despite extensive research efforts, the function of WT MYOC has remained elusive, and how mutant MYOC is linked to glaucoma is unclear. Mutant MYOC is believed to be misfolded within the endoplasmic reticulum, and under normal physiological conditions misfolded MYOC should be retro-translocated to the cytoplasm for degradation. To better understand mutant MYOC pathology, we CRISPR-engineered a rat to have a MYOC Y435H substitution that is the equivalent of the pathological human MYOC Y437H mutation. Using this engineered animal model, we discovered that the chaperone αB-crystallin (CRYAB) is a MYOC-binding partner and that co-expression of these two proteins increases protein aggregates. Our results suggest that the misfolded mutant MYOC aggregates with cytoplasmic CRYAB and thereby compromises protein clearance mechanisms in trabecular meshwork cells, and this process represents the primary mode of mutant MYOC pathology. We propose a model by which mutant MYOC causes glaucoma, and we propose that therapeutic treatment of patients having a MYOC mutation may focus on disrupting the MYOC-CRYAB complexes.
Collapse
Affiliation(s)
- Jeffrey M Lynch
- From Ophthalmology, Novartis Institutes for BioMedical Research, Cambridge, Massachusetts 02139.
| | - Bing Li
- From Ophthalmology, Novartis Institutes for BioMedical Research, Cambridge, Massachusetts 02139
| | - Parvaneh Katoli
- From Ophthalmology, Novartis Institutes for BioMedical Research, Cambridge, Massachusetts 02139
| | - Chuanxi Xiang
- From Ophthalmology, Novartis Institutes for BioMedical Research, Cambridge, Massachusetts 02139
| | - Barrett Leehy
- From Ophthalmology, Novartis Institutes for BioMedical Research, Cambridge, Massachusetts 02139
| | - Nalini Rangaswamy
- From Ophthalmology, Novartis Institutes for BioMedical Research, Cambridge, Massachusetts 02139
| | - Veronica Saenz-Vash
- Analytical Sciences and Imaging, Novartis Institutes for BioMedical Research, Cambridge, Massachusetts 02139
| | - Y Karen Wang
- Analytical Sciences and Imaging, Novartis Institutes for BioMedical Research, Cambridge, Massachusetts 02139
| | - Hong Lei
- Laboratory Animal Services, Novartis Institutes for BioMedical Research, Cambridge, Massachusetts 02139
| | - Thomas B Nicholson
- Chemical Biology and Therapeutics, and Novartis Institutes for BioMedical Research, Cambridge, Massachusetts 02139
| | - Erik Meredith
- Global Developmental Chemistry, Novartis Institutes for BioMedical Research, Cambridge, Massachusetts 02139
| | - Dennis S Rice
- From Ophthalmology, Novartis Institutes for BioMedical Research, Cambridge, Massachusetts 02139
| | - Ganesh Prasanna
- From Ophthalmology, Novartis Institutes for BioMedical Research, Cambridge, Massachusetts 02139
| | - Amy Chen
- From Ophthalmology, Novartis Institutes for BioMedical Research, Cambridge, Massachusetts 02139
| |
Collapse
|
19
|
Detection of mutations in MYOC, OPTN, NTF4, WDR36 and CYP1B1 in Chinese juvenile onset open-angle glaucoma using exome sequencing. Sci Rep 2018. [PMID: 29540704 PMCID: PMC5852028 DOI: 10.1038/s41598-018-22337-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Juvenile onset open-angle glaucoma (JOAG) affects patients before 40 years of age, causing high intraocular pressure and severe optic nerve damage. To expand the mutation spectrum of the causative genes in JOAG, with a view to identify novel disease-causing mutations, we investigated MYOC, OPTN, NTF4, WDR36 and CYP1B1 in a cohort of 67 unrelated Chinese JOAG patients. Whole exome sequencing was used to identify possible pathogenic mutations, which were further excluded in normal controls. After sequencing and the use of a database pipeline, as well as predictive assessment filtering, we identified a total of six mutations in three genes, MYOC, OPTN and CYP1B1. Among them, 2 heterozygous mutations in MYOC (c. 1109C > T, p. (P370L); c. 1150G > C, p. (D384H)), 2 heterozygous mutations in OPTN (c. 985A > G, p.(R329G); c. 1481T > G, p. (L494W)) and 2 homozygous mutations in CYP1B1 (c. 1412T > G, p.(I471S); c. 1169G > A, p.(R390H)) were identified as potentially causative mutations. No mutation was detected in NTF4 or WDR36. Our results enrich the mutation spectra and frequencies of MYOC, OPTN and CYP1B1 in JOAG among the Chinese population. Further studies are needed to address the pathogenicity of each of the mutations detected in this study.
Collapse
|
20
|
Chitranshi N, Dheer Y, Abbasi M, You Y, Graham SL, Gupta V. Glaucoma Pathogenesis and Neurotrophins: Focus on the Molecular and Genetic Basis for Therapeutic Prospects. Curr Neuropharmacol 2018; 16:1018-1035. [PMID: 29676228 PMCID: PMC6120108 DOI: 10.2174/1570159x16666180419121247] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Revised: 04/10/2018] [Accepted: 04/18/2018] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Retinal ganglion cell (RGC) degeneration is a major feature of glaucoma pathology. Neuroprotective approaches that delay or halt the progression of RGC loss are needed to prevent vision loss which can occur even after conventional medical or surgical treatments to lower intraocular pressure. OBJECTIVE The aim of this review was to examine the progress in genetics and cellular mechanisms associated with endoplasmic reticulum (ER) stress, RGC dysfunction and cell death pathways in glaucoma. MATERIALS AND METHODS Here, we review the involvement of neurotrophins like brain derived neurotrophic factor (BDNF) and its high affinity receptor tropomyosin receptor kinase (TrkB) in glaucoma. The role of ER stress markers in human and animal retinas in health and disease conditions is also discussed. Further, we analysed the literature highlighting genetic linkage in the context of primary open angle glaucoma and suggested mechanistic insights into potential therapeutic options relevant to glaucoma management. RESULTS The literature review of the neurobiology underlying neurotrophin pathways, ER stress and gene associations provide critical insights into association of RGCs death in glaucoma. Alteration in signalling pathway is associated with increased risk of misfolded protein aggregation in ER promoting RGC apoptosis. Several genes that are linked with neurotrophin signalling pathways have been reported to be associated with glaucoma pathology. CONCLUSION Understanding genetic heterogeneity and involvement of neurotrophin biology in glaucoma could help to understand the complex pathophysiology of glaucoma. Identification of novel molecular targets will be critical for drug development and provide neuroprotection to the RGCs and optic nerve.
Collapse
Affiliation(s)
- Nitin Chitranshi
- Address correspondence to this author at the Faculty of Medicine and Health Sciences, 75, Talavera Road, Macquarie University, Sydney, NSW 2109, Australia; Tel: +61-298502760; E-mail:
| | | | | | | | | | | |
Collapse
|
21
|
Saccà SC, Gandolfi S, Bagnis A, Manni G, Damonte G, Traverso CE, Izzotti A. From DNA damage to functional changes of the trabecular meshwork in aging and glaucoma. Ageing Res Rev 2016; 29:26-41. [PMID: 27242026 DOI: 10.1016/j.arr.2016.05.012] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 05/25/2016] [Accepted: 05/26/2016] [Indexed: 12/24/2022]
Abstract
Glaucoma is a degenerative disease of the eye. Both the anterior and posterior segments of the eye are affected, extensive damage being detectable in the trabecular meshwork and the inner retina-central visual pathway complex. Oxidative stress is claimed to be mainly responsible for molecular damage in the anterior chamber. Indeed, oxidation harms the trabecular meshwork, leading eventually to endothelial cell decay, tissue malfunction, subclinical inflammation, changes in the extracellular matrix and cytoskeleton, altered motility, reduced outflow facility and (ultimately) increased IOP. Moreover, free radicals are involved in aging and can be produced in the brain (as well as in the eye) as a result of ischemia, leading to oxidation of the surrounding neurons. Glaucoma-related cell death occurs by means of apoptosis, and apoptosis is triggered by oxidative stress via (a) mitochondrial damage, (b) inflammation, (c) endothelial dysregulation and dysfunction, and (d) hypoxia. The proteomics of the aqueous humor is significantly altered in glaucoma as a result of oxidation-induced trabecular damage. Those proteins whose aqueous humor levels are increased in glaucoma are biomarkers of trabecular meshwork impairment. Their diffusion from the anterior to the posterior segment of the eye may be relevant in the cascade of events triggering apoptosis in the inner retinal layers, including the ganglion cells.
Collapse
Affiliation(s)
- Sergio Claudio Saccà
- IRCCS San Martino University Hospital, Department of Neuroscience and Sense Organs, San Martino Hospital, Ophthalmology Unit, Viale Benedetto XV, 16132 Genoa, Italy.
| | - Stefano Gandolfi
- Ophthalmology Unit, Department of Biological, Biotechnological and Translational Sciences, University of Parma, Parma, Italy
| | - Alessandro Bagnis
- University of Genoa, Eye Clinic, Department of Neuroscience and Sense Organs, Viale Benedetto XV, 5, 16148 Genoa, Italy
| | - Gianluca Manni
- Dept. of Clinical Science and Translational Medicine, University Tor Vergata, Rome, Italy
| | - Gianluca Damonte
- Dept. of Experimental Medicine, Section of Biochemistry and Center of Excellence for Biomedical Research, University of Genoa, Viale Benedetto XV 1, 16132 Genoa, Italy
| | - Carlo Enrico Traverso
- University of Genoa, Eye Clinic, Department of Neuroscience and Sense Organs, Viale Benedetto XV, 5, 16148 Genoa, Italy
| | - Alberto Izzotti
- Mutagenesis Unit, IRCCS San Martino University Hospital, IST National Institute for Cancer Research, Department of Health Sciences, University of Genoa, Via A. Pastore 1, Genoa I-16132, Italy
| |
Collapse
|
22
|
Keller KE, Wirtz MK. Working your SOCS off: The role of ASB10 and protein degradation pathways in glaucoma. Exp Eye Res 2016; 158:154-160. [PMID: 27296073 DOI: 10.1016/j.exer.2016.06.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 06/01/2016] [Accepted: 06/07/2016] [Indexed: 12/25/2022]
Abstract
Evidence is accumulating to suggest that mutations in the Ankyrin and SOCS Box-containing protein-10 (ASB10) gene are associated with glaucoma. Since its identification in a large Oregon family with primary open-angle glaucoma (POAG), ASB10 variants have been associated with disease in US, German and Pakistani cohorts. ASB10 is a member of the ASB family of proteins, which have a common structure including a unique N-terminus, a variable number of central ankyrin (ANK) repeat domains and a suppressor of cytokine signaling (SOCS) box at the C-terminus. Mutations in ASB10 are distributed throughout the entire length of the gene including the two alternatively spliced variants of exon 1. A homozygous mutation in a Pakistani individual with POAG, which lies in the center of the SOCS box, is associated with a particularly severe form of the disease. Like other SOCS box-containing proteins, ASB10 functions in ubiquitin-mediated degradation pathways. The ANK repeats bind to proteins destined for degradation. The SOCS box recruits ubiquitin ligase proteins to form a complex to transfer ubiquitin to a substrate bound to the ANK repeats. The ubiquitin-tagged protein then enters either the proteasomal degradation pathway or the autophagic-lysosomal pathway. The choice of pathway appears to be dependent on which lysine residues are used to build polyubiquitin chains. However, these reciprocal pathways work in tandem to degrade proteins because inhibition of one pathway increases degradation via the other pathway. In this publication, we will review the literature that supports identification of ASB10 as a glaucoma-associated gene and the current knowledge of the function of the ASB10 protein. In addition, we present new data that indicates ASB10 expression is up-regulated by the inflammatory cytokines tumor necrosis factor-α and interleukin-1α. Finally, we will describe the emerging role of other SOCS box-containing proteins in protein degradation pathways in ocular cells.
Collapse
Affiliation(s)
- Kate E Keller
- Casey Eye Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA.
| | - Mary K Wirtz
- Casey Eye Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| |
Collapse
|
23
|
Yang X, Hondur G, Li M, Cai J, Klein JB, Kuehn MH, Tezel G. Proteomics Analysis of Molecular Risk Factors in the Ocular Hypertensive Human Retina. Invest Ophthalmol Vis Sci 2015; 56:5816-30. [PMID: 26348630 DOI: 10.1167/iovs.15-17294] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE To better understand ocular hypertension-induced early molecular alterations that may determine the initiation of neurodegeneration in human glaucoma, this study analyzed retinal proteomic alterations in the ocular hypertensive human retina. METHODS Retina samples were obtained from six human donors with ocular hypertension (without glaucomatous injury) and six age- and sex-matched normotensive controls. Retinal proteins were analyzed by two-dimensional LC-MS/MS (liquid chromatography and linear ion trap mass spectrometry) using oxygen isotope labeling for relative quantification of protein expression. Proteomics data were validated by Western blot and immunohistochemical analyses of selected proteins. RESULTS Out of over 2000 retinal proteins quantified, hundreds exhibited over 2-fold increased or decreased expression in ocular hypertensive samples relative to normotensive controls. Bioinformatics linked the proteomics datasets to various pathways important for maintenance of cellular homeostasis in the ocular hypertensive retina. Upregulated proteins included various heat shock proteins, ubiquitin proteasome pathway components, antioxidants, and DNA repair enzymes, while many proteins involved in mitochondrial oxidative phosphorylation exhibited downregulation in the ocular hypertensive retina. Despite the altered protein expression reflecting intrinsic adaptive/protective responses against mitochondrial energy failure, oxidative stress, and unfolded proteins, no alterations suggestive of an ongoing cell death process or neuroinflammation were detectable. CONCLUSIONS This study provides information about ocular hypertension-related molecular risk factors for glaucoma development. Molecular alterations detected in the ocular hypertensive human retina as opposed to previously detected alterations in human donor retinas with clinically manifest glaucoma suggest that proteome alterations determine the individual threshold to tolerate the ocular hypertension-induced tissue stress or convert to glaucomatous neurodegeneration when intrinsic adaptive/protective responses are overwhelmed.
Collapse
Affiliation(s)
- Xiangjun Yang
- Department of Ophthalmology Columbia University College of Physicians and Surgeons, New York, New York, United States
| | - Gözde Hondur
- Department of Ophthalmology Columbia University College of Physicians and Surgeons, New York, New York, United States
| | - Ming Li
- Department of Medicine, University of Louisville School of Medicine, Louisville, Kentucky, United States
| | - Jian Cai
- Department of Medicine, University of Louisville School of Medicine, Louisville, Kentucky, United States
| | - Jon B Klein
- Department of Medicine, University of Louisville School of Medicine, Louisville, Kentucky, United States 3Robley Rex Veterans Administration Medical Center, Louisville, Kentucky, United States
| | - Markus H Kuehn
- Department of Ophthalmology & Visual Sciences, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States
| | - Gülgün Tezel
- Department of Ophthalmology Columbia University College of Physicians and Surgeons, New York, New York, United States
| |
Collapse
|
24
|
Genes, pathways, and animal models in primary open-angle glaucoma. Eye (Lond) 2015; 29:1285-98. [PMID: 26315706 DOI: 10.1038/eye.2015.160] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 07/27/2015] [Indexed: 02/08/2023] Open
Abstract
Glaucoma is an optic neuropathy characterized by loss of retinal ganglion cells (RGCs) and consequently visual field loss. It is a complex and heterogeneous disease in which both environmental and genetic factors play a role. With the advent of genome-wide association studies (GWASs), the number of loci associated with primary open-angle glaucoma (POAG) have increased greatly. There has also been major progress in understanding the genes determining the vertical cup-disc ratio (VCDR), disc area (DA), cup area (CA), intraocular pressure (IOP), and central corneal thickness (CCT). In this review, we will update and summarize the genetic loci associated so far with POAG, VCDR, DA, CA, IOP, and CCT. We will describe the pathways revealed and supported by genetic association studies, integrating current knowledge from human and experimental data. Finally, we will discuss approaches for functional genomics and clinical translation.
Collapse
|
25
|
Liton PB. The autophagic lysosomal system in outflow pathway physiology and pathophysiology. Exp Eye Res 2015; 144:29-37. [PMID: 26226231 DOI: 10.1016/j.exer.2015.07.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 07/06/2015] [Accepted: 07/22/2015] [Indexed: 12/19/2022]
Abstract
Malfunction of the trabecular meshwork (TM)/schlemm's canal (SC) conventional outflow pathway is associated with elevated intraocular pressure (IOP) and, therefore, increased risk of developing glaucoma, a potentially blinding disease affecting more than 70 million people worldwide. This TM/SC tissue is subjected to different types of stress, including mechanical, oxidative, and phagocytic stress. Long-term exposure to these stresses is believed to lead to a progressive accumulation of damaged cellular and tissue structures causing permanent alterations in the tissue physiology, and contribute to the pathologic increase in aqueous humor (AH) outflow resistance. Autophagy is emerging as an essential cellular survival mechanism against a variety of stressors. In addition to performing basal functions, autophagy acts as a cellular survival pathway and represents an essential mechanism by which organisms can adapt to acute stress conditions and repair stress-induced damage. A decline in autophagy has been observed in most tissues with aging and has been considered responsible, at least in part, for the accumulation of damaged cellular components in almost all tissues of aging organisms. Dysfunction in the autophagy pathway is associated with several human diseases, from infectious diseases to cancer and neurodegeneration. In this review, we will summarize our current knowledge of the emerging roles of autophagy in outflow tissue physiology and pathophysiology, including novel evidence suggesting compromised autophagy in the glaucomatous outflow pathway.
Collapse
Affiliation(s)
- Paloma B Liton
- Duke University, Department of Ophthalmology, Durham, NC, USA.
| |
Collapse
|
26
|
Porter K, Hirt J, Stamer WD, Liton PB. Autophagic dysregulation in glaucomatous trabecular meshwork cells. Biochim Biophys Acta Mol Basis Dis 2014; 1852:379-85. [PMID: 25483712 DOI: 10.1016/j.bbadis.2014.11.021] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Revised: 11/12/2014] [Accepted: 11/30/2014] [Indexed: 01/07/2023]
Abstract
Primary open angle glaucoma (POAG) is a degenerative disease commonly associated with aging and elevated intraocular pressure (IOP). Higher resistance to aqueous humor (AH) outflow through the trabecular meshwork (TM) generates the elevated IOP in POAG; unfortunately the underlying molecular mechanisms responsible for elevated resistance are unknown. It is widely accepted, however, that differences between normal and POAG TM tissues are presumably a consequence of cellular dysfunction. Here, we investigated the autophagic function and response to chronic oxidative stress in TM cells isolated from glaucomatous and age-matched donor eyes. Glaucomatous TM cells showed elevated senescence-associated-beta-galactosidase (SA-β-Gal) and cellular lipofuscin, together with decreased steady-state levels of LC3B-II, decreased levels of pRPS6K-T389 and reduced proteolysis of long-live proteins. Moreover, the glaucomatous cultures failed to activate autophagy when exposed to hyperoxic conditions. These results strongly suggest mTOR-dependent dysregulation of the autophagic pathway in cells isolated from the glaucomatous TM. Such dysregulated autophagic capacity can have a detrimental impact in outflow pathway tissue, i.e. mechanotransduction, and thus represent an important factor contributing to the progression of the disease.
Collapse
Affiliation(s)
- Kristine Porter
- Duke University, Department of Ophthalmology, Durham, NC, USA
| | - Joshua Hirt
- Duke University, Department of Ophthalmology, Durham, NC, USA
| | - W Daniel Stamer
- Duke University, Department of Ophthalmology, Durham, NC, USA
| | - Paloma B Liton
- Duke University, Department of Ophthalmology, Durham, NC, USA.
| |
Collapse
|
27
|
Faralli JA, Clark RW, Filla MS, Peters DM. NFATc1 activity regulates the expression of myocilin induced by dexamethasone. Exp Eye Res 2014; 130:9-16. [PMID: 25450062 DOI: 10.1016/j.exer.2014.11.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 10/24/2014] [Accepted: 11/17/2014] [Indexed: 10/24/2022]
Abstract
Mutations in the myocilin gene (MYOC) account for 10% of juvenile open-angle glaucoma cases and 3-4% of adult onset primary open-angle glaucoma cases. It is a secreted glycoprotein found in many ocular and non-ocular tissues and has been linked to elevated intraocular pressure. In human trabecular meshwork (HTM) cells, MYOC expression can be induced by the glucocorticoid dexamethasone (DEX). In this study we examined the role of the calcineurin/NFATc1 (Nuclear Factor of Activated T-cells) pathway in the DEX induction of MYOC in HTM cells. In post-confluent HTM cells treated with either 500 nM DEX or 0.1% ethanol (EtOH; vehicle control) for 0-6 days both protein and mRNA levels of MYOC were increased while DEX was present. The protein and mRNA levels remained elevated for an additional 12 days after the removal of DEX. Only 1 day of DEX treatment was sufficient to trigger a sustained increase in MYOC mRNA that lasted for 4 days after the removal of DEX. Similar to other studies, myocilin protein expression was not seen until the second day of DEX treatment while mRNA increased within one day of DEX indicating that this is a secondary glucocorticoid response. To determine if MYOC gene expression was regulated by calcineurin/NFATc1, HTM cells were pre-treated for 1 h with the calcineurin inhibitors cyclosporin A or INCA-6 prior to the addition of DEX or EtOH for 2 days. NFATc1 siRNA was used to determine if NFATc1 was required for MYOC mRNA expression. Cells were also treated with the ionophone ionomycin to determine if increased cytosolic calcium affected MYOC expression. These studies showed that the DEX induced increase in MYOC mRNA could be inhibited with either cyclosporin A or INCA-6 or by transfection with NFATc1 siRNA and that ionomycin was unable to increase MYOC mRNA. Immunofluorescence microscopy was also performed to determine if DEX caused the nuclear translocation of NFATc1. Immunostaining showed that NFATc1 relocated to the nucleus within 15 min of DEX treatment and remained there for up to 2 h. The data suggest that the DEX-induced increase in MYOC expression activates a calcineurin and NFATc1 pathway in a calcium independent mechanism.
Collapse
Affiliation(s)
- Jennifer A Faralli
- Departments of Pathology & Laboratory Medicine, University of Wisconsin, Madison, WI 53706, USA.
| | - Ross W Clark
- Departments of Pathology & Laboratory Medicine, University of Wisconsin, Madison, WI 53706, USA
| | - Mark S Filla
- Departments of Ophthalmology & Visual Sciences, University of Wisconsin, Madison, WI 53706, USA
| | - Donna M Peters
- Departments of Pathology & Laboratory Medicine, University of Wisconsin, Madison, WI 53706, USA; Departments of Ophthalmology & Visual Sciences, University of Wisconsin, Madison, WI 53706, USA
| |
Collapse
|