1
|
Rognon GT, Liao AYA, Pasteurin RP, Soundararajan A, Pattabiraman PP. Lipids and lipid regulators in intraocular pressure homeostasis. Curr Opin Pharmacol 2025; 82:102523. [PMID: 40245644 DOI: 10.1016/j.coph.2025.102523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 02/12/2025] [Accepted: 03/13/2025] [Indexed: 04/19/2025]
Abstract
Increased intraocular pressure is the strongest correlated modifiable risk factor for developing primary open-angle glaucoma (POAG). Lipids have long been known to be a major constituent of aqueous humor. Lipid mediators, prostaglandins for example, are the first-line treatment for glaucoma. Innovative technologies have made the investigation of lipids in small quantities possible, and interest in identifying lipids as new pharmacological targets has grown in ophthalmology. There is expanding evidence to suggest that lipids and their active metabolites play a role in POAG pathophysiology, as differences between control and diseased eyes have now been demonstrated. The role of these differences is yet to be determined and is the subject of this review.
Collapse
Affiliation(s)
- Gregory T Rognon
- Glick Eye Institute, Department of Ophthalmology, Indiana University School of Medicine, 1160 West Michigan Street, Indianapolis, IN, 46202-5209, USA
| | - Anna Yu-An Liao
- Glick Eye Institute, Department of Ophthalmology, Indiana University School of Medicine, 1160 West Michigan Street, Indianapolis, IN, 46202-5209, USA; Carmel High School, 520 E Main St, Carmel, IN, 46032, USA
| | - Rodahina Philihina Pasteurin
- Glick Eye Institute, Department of Ophthalmology, Indiana University School of Medicine, 1160 West Michigan Street, Indianapolis, IN, 46202-5209, USA
| | - Avinash Soundararajan
- Glick Eye Institute, Department of Ophthalmology, Indiana University School of Medicine, 1160 West Michigan Street, Indianapolis, IN, 46202-5209, USA
| | - Padmanabhan Paranji Pattabiraman
- Glick Eye Institute, Department of Ophthalmology, Indiana University School of Medicine, 1160 West Michigan Street, Indianapolis, IN, 46202-5209, USA; Stark Neuroscience Research Institute, Indiana University School of Medicine, 320 W. 15th Street, Indiana, 46202-5209, USA.
| |
Collapse
|
2
|
Ghosh R, Herberg S. The role of YAP/TAZ mechanosignaling in trabecular meshwork and Schlemm's canal cell dysfunction. Vision Res 2024; 224:108477. [PMID: 39208753 PMCID: PMC11470804 DOI: 10.1016/j.visres.2024.108477] [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: 06/01/2024] [Revised: 08/22/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024]
Abstract
This focused review highlights the importance of yes-associated protein (YAP)/transcriptional coactivator with PDZ binding motif (TAZ) mechanosignaling in human trabecular meshwork and Schlemm's canal cells in response to glaucoma-associated extracellular matrix stiffening and cyclic mechanical stretch, as well as biochemical pathway modulators (with signaling crosstalk) including transforming growth factor beta 2, glucocorticoids, Wnt, lysophosphatidic acid, vascular endothelial growth factor, and oxidative stress. We provide a comprehensive overview of relevant literature from the last decade, highlight intriguing research avenues with translational potential, and close with an outlook on future directions.
Collapse
Affiliation(s)
- Rajanya Ghosh
- Department of Ophthalmology and Visual Sciences, Center for Vision Research, SUNY Upstate Medical University, Syracuse, NY 13210, USA; Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Samuel Herberg
- Department of Ophthalmology and Visual Sciences, Center for Vision Research, SUNY Upstate Medical University, Syracuse, NY 13210, USA; Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA; Department of Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA; BioInspired Institute, Syracuse University, Syracuse, NY 13244, USA; Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, NY 13244, USA.
| |
Collapse
|
3
|
Mathew DJ, Sivak JM. Lipid mediators in glaucoma: Unraveling their diverse roles and untapped therapeutic potential. Prostaglandins Other Lipid Mediat 2024; 171:106815. [PMID: 38280539 DOI: 10.1016/j.prostaglandins.2024.106815] [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: 08/25/2023] [Revised: 01/19/2024] [Accepted: 01/23/2024] [Indexed: 01/29/2024]
Abstract
Glaucoma is a complex neurodegenerative disease characterized by optic nerve damage and visual field loss, and remains a leading cause of irreversible blindness. Elevated intraocular pressure (IOP) is a critical risk factor that requires effective management. Emerging research underscores dual roles of bioactive lipid mediators in both IOP regulation, and the modulation of neurodegeneration and neuroinflammation in glaucoma. Bioactive lipids, encompassing eicosanoids, specialized pro-resolving mediators (SPMs), sphingolipids, and endocannabinoids, have emerged as crucial players in these processes, orchestrating inflammation and diverse effects on aqueous humor dynamics and tissue remodeling. Perturbations in these lipid mediators contribute to retinal ganglion cell loss, vascular dysfunction, oxidative stress, and neuroinflammation. Glaucoma management primarily targets IOP reduction via pharmacological agents and surgical interventions, with prostaglandin analogues at the forefront. Intriguingly, additional lipid mediators offer promise in attenuating inflammation and providing neuroprotection. Here we explore these pathways to shed light on their intricate roles, and to unveil novel therapeutic avenues for glaucoma management.
Collapse
Affiliation(s)
- D J Mathew
- Donald K Johnson Eye Institute, Krembil Research Institute, University Health Network, Toronto, Canada; Department of Ophthalmology and Vision Science, University of Toronto School of Medicine, Toronto, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto School of Medicine, Toronto, Canada
| | - J M Sivak
- Donald K Johnson Eye Institute, Krembil Research Institute, University Health Network, Toronto, Canada; Department of Ophthalmology and Vision Science, University of Toronto School of Medicine, Toronto, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto School of Medicine, Toronto, Canada.
| |
Collapse
|
4
|
Wang T, Soundararajan A, Rabinowitz J, Jaiswal A, Osborne T, Pattabiraman PP. Identification of the novel role of sterol regulatory element binding proteins (SREBPs) in mechanotransduction and intraocular pressure regulation. FASEB J 2023; 37:e23248. [PMID: 37823226 PMCID: PMC10826798 DOI: 10.1096/fj.202301185r] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 09/11/2023] [Accepted: 09/26/2023] [Indexed: 10/13/2023]
Abstract
Trabecular meshwork (TM) cells are contractile and mechanosensitive, and they aid in maintaining intraocular pressure (IOP) homeostasis. Lipids are attributed to modulating TM contractility, with poor mechanistic understanding. In this study using human TM cells, we identify the mechanosensing role of the transcription factors sterol regulatory element binding proteins (SREBPs) involved in lipogenesis. By constitutively activating SREBPs and pharmacologically inactivating SREBPs, we have mechanistically deciphered the attributes of SREBPs in regulating the contractile properties of TM. The pharmacological inhibition of SREBPs by fatostatin and molecular inactivation of SREBPs ex vivo and in vivo, respectively, results in significant IOP lowering. As a proof of concept, fatostatin significantly decreased the SREBPs responsive genes and enzymes involved in lipogenic pathways as well as the levels of the phospholipid, cholesterol, and triglyceride. Further, we show that fatostatin mitigated actin polymerization machinery and stabilization, and decreased ECM synthesis and secretion. We thus postulate that lowering lipogenesis in the TM outflow pathway can hold the key to lowering IOP by modifying the TM biomechanics.
Collapse
Affiliation(s)
- Ting Wang
- Glick Eye Institute, Department of Ophthalmology, Indiana University School of Medicine, 1160 West Michigan Street, Indianapolis, Indiana, 46202, United States of America
- Stark Neuroscience Research Institute, Medical Neuroscience Graduate Program, Indiana University School of Medicine, 320 W. 15th Street, Indiana, 46202, United States of America
| | - Avinash Soundararajan
- Glick Eye Institute, Department of Ophthalmology, Indiana University School of Medicine, 1160 West Michigan Street, Indianapolis, Indiana, 46202, United States of America
| | - Jeffrey Rabinowitz
- Department of Ophthalmology, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Anant Jaiswal
- Institute for Fundamental Biomedical Research, Department of Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, St. Petersburg, Florida, 33701, United States of America
| | - Timothy Osborne
- Institute for Fundamental Biomedical Research, Department of Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, St. Petersburg, Florida, 33701, United States of America
| | - Padmanabhan Paranji Pattabiraman
- Glick Eye Institute, Department of Ophthalmology, Indiana University School of Medicine, 1160 West Michigan Street, Indianapolis, Indiana, 46202, United States of America
- Stark Neuroscience Research Institute, Medical Neuroscience Graduate Program, Indiana University School of Medicine, 320 W. 15th Street, Indiana, 46202, United States of America
| |
Collapse
|
5
|
Wang T, Soundararajan A, Rabinowitz J, Jaiswal A, Osborne T, Pattabiraman PP. Identification of the novel role of sterol regulatory element binding proteins (SREBPs) in mechanotransduction and intraocular pressure regulation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.05.527136. [PMID: 37214961 PMCID: PMC10197526 DOI: 10.1101/2023.02.05.527136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Trabecular meshwork (TM) cells are highly contractile and mechanosensitive to aid in maintaining intraocular pressure (IOP) homeostasis. Lipids are attributed to modulating TM contractility with poor mechanistic understanding. In this study using human TM cells, we identify the mechanosensing role of the transcription factors sterol regulatory element binding proteins (SREBPs) involved in lipogenesis. By constitutively activating SREBPs and pharmacologically inactivating SREBPs, we have mechanistically deciphered the attributes of SREBPs in regulating the contractile properties of TM. The pharmacological inhibition of SREBPs by fatostatin and molecular inactivation of SREBPs ex vivo and in vivo respectively results in significant IOP lowering. As a proof of concept, fatostatin significantly decreased the SREBPs responsive genes and enzymes involved in lipogenic pathways as well as the levels of the phospholipid, cholesterol, and triglyceride. Further, we show that fatostatin mitigated actin polymerization machinery and stabilization, and decreased ECM synthesis and secretion. We thus postulate that lowering lipogenesis in the TM outflow pathway can hold the key to lowering IOP by modifying the TM biomechanics. Synopsis In this study, we show the role of lipogenic transcription factors sterol regulatory element binding proteins (SREBPs) in the regulation of intraocular pressure (IOP). ( Synopsis Figure - Created using Biorender.com ) SREBPs are involved in the sensing of changes in mechanical stress on the trabecular meshwork (TM). SREBPs aid in transducing the mechanical signals to induce actin polymerization and filopodia/lamellipodia formation.SREBPs inactivation lowered genes and enzymes involved in lipogenesis and modified lipid levels in TM.SREBPs activity is a critical regulator of ECM engagement to the matrix sites.Inactivation of SCAP-SREBP pathway lowered IOP via actin relaxation and decreasing ECM production and deposition in TM outflow pathway signifying a novel relationship between SREBP activation status and achieving IOP homeostasis.
Collapse
|
6
|
Soundararajan A, Wang T, Sundararajan R, Wijeratne A, Mosley A, Harvey FC, Bhattacharya S, Pattabiraman PP. Multiomics analysis reveals the mechanical stress-dependent changes in trabecular meshwork cytoskeletal-extracellular matrix interactions. Front Cell Dev Biol 2022; 10:874828. [PMID: 36176278 PMCID: PMC9513235 DOI: 10.3389/fcell.2022.874828] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 08/12/2022] [Indexed: 11/29/2022] Open
Abstract
Trabecular meshwork (TM) tissue is subjected to constant mechanical stress due to the ocular pulse created by the cardiac cycle. This brings about alterations in the membrane lipids and associated cell-cell adhesion and cell-extracellular matrix (ECM) interactions, triggering intracellular signaling responses to counter mechanical insults. A loss of such response can lead to elevated intraocular pressure (IOP), a major risk factor for primary open-angle glaucoma. This study is aimed to understand the changes in signaling responses by TM subjected to mechanical stretch. We utilized multiomics to perform an unbiased mRNA sequencing to identify changes in transcripts, mass spectrometry- (MS-) based quantitative proteomics for protein changes, and multiple reaction monitoring (MRM) profiling-based MS and high-performance liquid chromatography (HPLC-) based MS to characterize the lipid changes. We performed pathway analysis to obtain an integrated map of TM response to mechanical stretch. The human TM cells subjected to mechanical stretch demonstrated an upregulation of protein quality control, oxidative damage response, pro-autophagic signal, induction of anti-apoptotic, and survival signaling. We propose that mechanical stretch-induced lipid signaling via increased ceramide and sphingomyelin potentially contributes to increased TM stiffness through actin-cytoskeleton reorganization and profibrotic response. Interestingly, increased phospholipids and diacylglycerol due to mechanical stretch potentially enable cell membrane remodeling and changes in signaling pathways to alter cellular contractility. Overall, we propose the mechanistic interplay of macromolecules to bring about a concerted cellular response in TM cells to achieve mechanotransduction and IOP regulation when TM cells undergo mechanical stretch.
Collapse
Affiliation(s)
- Avinash Soundararajan
- Department of Ophthalmology, Glick Eye Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Ting Wang
- Department of Ophthalmology, Glick Eye Institute, Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Rekha Sundararajan
- Department of Ophthalmology, Glick Eye Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Aruna Wijeratne
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, United States
- Center for Proteome Analysis, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Amber Mosley
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, United States
- Center for Proteome Analysis, Indiana University School of Medicine, Indianapolis, IN, United States
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Faith Christine Harvey
- Bascom Palmer Eye Institute, Miller School of Medicine at University of Miami, Miami, FL, United States
- Miami Integrative Metabolomics Research Center, Miami, FL, United States
| | - Sanjoy Bhattacharya
- Bascom Palmer Eye Institute, Miller School of Medicine at University of Miami, Miami, FL, United States
- Miami Integrative Metabolomics Research Center, Miami, FL, United States
| | - Padmanabhan Paranji Pattabiraman
- Department of Ophthalmology, Glick Eye Institute, Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
7
|
Fibrotic Response of Human Trabecular Meshwork Cells to Transforming Growth Factor-Beta 3 and Autotaxin in Aqueous Humor. Biomolecules 2022; 12:biom12091231. [PMID: 36139071 PMCID: PMC9496180 DOI: 10.3390/biom12091231] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 08/28/2022] [Accepted: 09/01/2022] [Indexed: 11/16/2022] Open
Abstract
This study examines the potential role of transforming growth factor-beta 3 (TGF-β3) on the fibrotic response of cultured human trabecular meshwork (HTM) cells. The relationships and trans-signaling interactions between TGF-β3 and autotaxin (ATX) in HTM cells were also examined. The levels of TGF-β and ATX in the aqueous humor (AH) of patients were measured by an immunoenzymetric assay. The TGF-β3-induced expression of the fibrogenic markers, fibronectin, collagen type I alpha 1 chain, and alpha-smooth muscle actin, and ATX were examined by quantitative real-time PCR, Western blotting, and immunocytochemistry, and the trans-signaling regulatory effect of TGF-β3 on ATX expression was also evaluated. In HTM cells, the significant upregulation of ATX was induced by TGF-β3 at a concentration of 0.1 ng/mL, corresponding to the physiological concentration in the AH of patients with exfoliative glaucoma (XFG). However, higher concentrations of TGF-β3 significantly suppressed ATX expression. TGF-β3 regulated ATX transcription and signaling in HTM cells, inducing the upregulation of fibrogenic proteins in a dose-dependent manner. Trans-signaling of TGF-β3 regulated ATX transcription, protein expression, and signaling, and was thereby suggested to induce fibrosis of the trabecular meshwork. Modulation of trans-signaling between TGF-β3 and ATX may be key to elucidate the pathology of XFG, and for the development of novel treatment modalities.
Collapse
|
8
|
Uchida T, Shimizu S, Yamagishi R, Tokuoka SM, Kita Y, Sakata R, Honjo M, Aihara M. TRPV4 is activated by mechanical stimulation to induce prostaglandins release in trabecular meshwork, lowering intraocular pressure. PLoS One 2021; 16:e0258911. [PMID: 34673834 PMCID: PMC8530296 DOI: 10.1371/journal.pone.0258911] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 10/07/2021] [Indexed: 11/25/2022] Open
Abstract
Trabecular meshwork constitutes the conventional outflow pathway and controls intraocular pressure by regulating aqueous outflow. Mechanical stimulation has been studied as one of the triggers to regulate aqueous outflow in trabecular meshwork, but it is not well understood. We investigated that how transient receptor potential cation channel subfamily V member 4 (TRPV4) functions in human trabecular meshwork cells (HTMC) and affects intraocular pressure (IOP). HTMC were treated with TRPV4 siRNA, followed by incubation for 24 hours. We confirmed the suppression of TRPV4 mRNA expression and the reduction of Ca2+ influx by the TRPV4 agonist GSK1016790A in TRPV4 siRNA-treated HTMC. TRPV4 siRNA-treated HTMC exhibited a significant reduction in Ca2+ influx and production of arachidonic acid and prostaglandin (PG) E2 induced by mechanical stretch, and direct activation of TRPV4 by GSK1016790A increased production of arachidonic acid, PGE2, and PGD2 and inhibited gel contraction. Furthermore, TRPV4-deficient mice had higher IOP than wild-type mice, and GSK1016790A administration lowered IOP. These results suggest that TRPV4 mediates the cellular response induced by trabecular meshwork stretch, leading to IOP reduction through the production of prostaglandins and inhibition of cell contraction. Targeting TRPV4 may have therapeutic benefits that lead to lowering IOP in glaucoma patients.
Collapse
Affiliation(s)
- Takatoshi Uchida
- Department of Ophthalmology, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
- Senju Laboratory of Ocular Science, Senju Pharmaceutical Co., Ltd., Kobe, Japan
| | - Shota Shimizu
- Department of Ophthalmology, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
- Senju Laboratory of Ocular Science, Senju Pharmaceutical Co., Ltd., Kobe, Japan
| | - Reiko Yamagishi
- Department of Ophthalmology, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
| | - Suzumi M. Tokuoka
- Department of Lipidomics, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
| | - Yoshihiro Kita
- Department of Lipidomics, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
- Life Science Core Facility, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
| | - Rei Sakata
- Department of Ophthalmology, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
| | - Megumi Honjo
- Department of Ophthalmology, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
| | - Makoto Aihara
- Department of Ophthalmology, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
- * E-mail:
| |
Collapse
|
9
|
Critical Roles of Lysophospholipid Receptors in Activation of Neuroglia and Their Neuroinflammatory Responses. Int J Mol Sci 2021; 22:ijms22157864. [PMID: 34360625 PMCID: PMC8346064 DOI: 10.3390/ijms22157864] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/21/2021] [Accepted: 07/21/2021] [Indexed: 12/12/2022] Open
Abstract
Activation of microglia and/or astrocytes often releases proinflammatory molecules as critical pathogenic mediators that can promote neuroinflammation and secondary brain damages in diverse diseases of the central nervous system (CNS). Therefore, controlling the activation of glial cells and their neuroinflammatory responses has been considered as a potential therapeutic strategy for treating neuroinflammatory diseases. Recently, receptor-mediated lysophospholipid signaling, sphingosine 1-phosphate (S1P) receptor- and lysophosphatidic acid (LPA) receptor-mediated signaling in particular, has drawn scientific interest because of its critical roles in pathogenies of diverse neurological diseases such as neuropathic pain, systemic sclerosis, spinal cord injury, multiple sclerosis, cerebral ischemia, traumatic brain injury, hypoxia, hydrocephalus, and neuropsychiatric disorders. Activation of microglia and/or astrocytes is a common pathogenic event shared by most of these CNS disorders, indicating that lysophospholipid receptors could influence glial activation. In fact, many studies have reported that several S1P and LPA receptors can influence glial activation during the pathogenesis of cerebral ischemia and multiple sclerosis. This review aims to provide a comprehensive framework about the roles of S1P and LPA receptors in the activation of microglia and/or astrocytes and their neuroinflammatory responses in CNS diseases.
Collapse
|
10
|
Igarashi N, Honjo M, Yamagishi R, Kurano M, Yatomi Y, Igarashi K, Kaburaki T, Aihara M. Crosstalk between transforming growth factor β-2 and Autotaxin in trabecular meshwork and different subtypes of glaucoma. J Biomed Sci 2021; 28:47. [PMID: 34140021 PMCID: PMC8212476 DOI: 10.1186/s12929-021-00745-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 06/11/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Elevated transforming growth factor (TGF)-β2 in aqueous humor (AH) has been suggested to contribute to trabecular meshwork (TM) fibrosis and intraocular pressure (IOP) regulation in primary open-angle glaucoma (POAG), but TGF-β2 is downregulated in secondary open-angle glaucoma (SOAG). Because autotaxin (ATX) is upregulated in SOAG, we investigated the relationships and trans-signaling interactions of these mediators. METHODS The level of ATX in AH was determined using a two-site immunoenzymetric assay, and TGF-β levels were measured using the Bio-Plex Pro TGF-β Assay. RNA scope was used to assess the expression of ATX and TGF-β2 in human's eye specimen. And in vitro studies were performed using hTM cells to explore if trans-signaling of TGF-β2 regulates ATX expressions. RESULTS TGF-β2/ATX ratio was significantly high in AH of control or POAG compared with SOAG, and negatively correlated with IOP. RNA scope revelated positive expressions of both TGF-β2 and ATX in ciliary body (CB) and TM in control, but ATX expressions was significantly enhanced in SOAG. In hTM cells, ATX expressions were regulated by TGF-β2 with concentration-dependent manner. In counter, ATX also induced TGF-β1, TGF-β2 and TGFBI upregulations and activation of the Smad-sensitive promoter, as well as upregulation of fibrotic markers, and these upregulation was significantly suppressed by both TGF-β and ATX inhibition. CONCLUSIONS Trans-signaling of TGF-β2 regulates ATX expressions and thereby induced upregulations of TGF-βs or fibrosis of hTM. TGF-β2 trans-signaling potently regulate ATX transcription and signaling in hTM cells, which may reflect different profile of these mediators in glaucoma subtypes. Trial Registration This prospective observational study was approved by the Institutional Review Board of the University of Tokyo and was registered with the University Hospital Medical Information Network Clinical Trials Registry of Japan (ID: UMIN000027137). All study procedures conformed to the Declaration of Helsinki. Written informed consent was obtained from each patient.
Collapse
Affiliation(s)
- Nozomi Igarashi
- Department of Ophthalmology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Megumi Honjo
- Department of Ophthalmology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
| | - Reiko Yamagishi
- Department of Ophthalmology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Makoto Kurano
- Department of Clinical Laboratory Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo, 113-8655, Japan.,CREST, Japan Science and Technology Corporation (JST), Saitama, Japan
| | - Yutaka Yatomi
- Department of Clinical Laboratory Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo, 113-8655, Japan.,CREST, Japan Science and Technology Corporation (JST), Saitama, Japan.,Department of Clinical Laboratory, The University of Tokyo Hospital, Tokyo, Japan
| | - Koji Igarashi
- Bioscience Division, Reagent Development Department, AIA Research Group, TOSOH Corporation, Kanagawa, Japan
| | - Toshikatsu Kaburaki
- Department of Ophthalmology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Ophthalmology, Jichi Medical University Saitama Medical Center, Saitama, Japan
| | - Makoto Aihara
- Department of Ophthalmology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
11
|
O'Regan A, O'Brien CJ, Eivers SB. The lysophosphatidic acid axis in fibrosis: Implications for glaucoma. Wound Repair Regen 2021; 29:613-626. [PMID: 34009724 DOI: 10.1111/wrr.12929] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/13/2021] [Accepted: 04/28/2021] [Indexed: 12/27/2022]
Abstract
Glaucoma is a common progressive optic neuropathy that results in visual field defects and can lead to irreversible blindness. The pathophysiology of glaucoma involves dysregulated extracellular matrix remodelling in both the trabecular meshwork in the anterior chamber and in the lamina cribrosa of the optic nerve head. Fibrosis in these regions leads to raised intraocular pressure and retinal ganglion cell degeneration, respectively. Lysophosphatidic acid (LPA) is a bioactive lipid mediator which acts via six G-protein coupled receptors on the cell surface to activate intracellular pathways that promote cell proliferation, transcription and survival. LPA signalling has been implicated in both normal wound healing and pathological fibrosis. LPA enhances fibroblast proliferation, migration and contraction, and induces expression of pro-fibrotic mediators such as connective tissue growth factor. The LPA axis plays a major role in diseases such as idiopathic pulmonary fibrosis, where it has been identified as an important pharmacological target. In glaucoma, LPA is present in high levels in the aqueous humour, and its signalling has been found to increase resistance to aqueous humour outflow through altered trabecular meshwork cellular contraction and extracellular matrix deposition. LPA signalling may, therefore, also represent an attractive target for treatment of glaucoma. In this review we wish to describe the role of LPA and its related proteins in tissue fibrosis and glaucoma.
Collapse
Affiliation(s)
- Amy O'Regan
- UCD Clinical Research Centre, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Colm J O'Brien
- UCD Clinical Research Centre, Mater Misericordiae University Hospital, Dublin, Ireland.,Department of Ophthalmology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Sarah B Eivers
- UCD Clinical Research Centre, Mater Misericordiae University Hospital, Dublin, Ireland
| |
Collapse
|
12
|
Milbeck SM, Bhattacharya SK. Alteration in Lysophospholipids and Converting Enzymes in Glaucomatous Optic Nerves. Invest Ophthalmol Vis Sci 2021; 61:60. [PMID: 32602905 PMCID: PMC7415893 DOI: 10.1167/iovs.61.6.60] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Purpose To determine whether lysophospholipid (LPL) profiles and corresponding conversion enzymes in the LPL pathways are altered in the optic nerve (ON) between human control and glaucoma samples. Methods Lipids extracted from control (n = 11) and glaucomatous (n = 12) ON samples using the Bligh and Dyer method were subjected to high-resolution mass spectrometry on a Q-exactive mass spectrometer coupled with a high-performance liquid chromatography (Accela 600) system. Analysis was performed for LPLs (lysophosphatidylcholines, lysophosphatidylserines, lysophosphatidylethanolamines, lysophosphatidylinositols, and lysosphingomyelines) using LipidSearch v.4.1, MZmine v.2.0, and MetaboAnalyst v.4.0. LPL synthesis and degradation pathway maps, utilizing UniProt and BRENDA database entries as needed, were created using Kyoto Encyclopedia of Genes and Genomes (KEGG)-based tools. The mRNA expression level in normal and glaucomatous human ON were analyzed using Gene Expression Omnibus (GEO) entry GSE45570. Protein amounts were determined using PHAST gel and dot blot and were used for normalization of protein amounts across samples. Western blot, ELISA, and protein quantification were performed using established protocols. Results Principal component analysis of ON LPL profile placed control and glaucomatous ONs in two distinct separate groups. Mass spectrometric analysis of ON revealed decrease in lysophosphatidic acid, lysophosphatidylethanolamine, lysophosphatidylcholine, and significant increase in diacylglycerol in glaucomatous ON. Statistical analysis of LPL conversion enzymes revealed significant overexpression of phosphatidate phosphatase LPIN2, phospholipid phosphatase 3, phosphatidylcholine-sterol acyltransferase, and calcium-dependent phospholipase 2, and significant downregulation of glycerol-3-phosphate acyltransferase 4 at mRNA level in glaucomatous ON. Western blot and ELISA confirmed proteomic differences between normal and diseased ON. Conclusions Our analysis revealed alterations in specific LPL levels and corresponding select enzyme-level changes in glaucomatous ON.
Collapse
|
13
|
Edwards G, Arcuri J, Wang H, Ziebarth N, Zode G, Lee RK, Bhattacharya SK. Endogenous ocular lipids as potential modulators of intraocular pressure. J Cell Mol Med 2020; 24:3856-3900. [PMID: 32090468 PMCID: PMC7171415 DOI: 10.1111/jcmm.14975] [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: 09/09/2019] [Revised: 12/04/2019] [Accepted: 12/21/2019] [Indexed: 12/13/2022] Open
Abstract
Elevated intraocular pressure (IOP) is a risk factor in glaucoma, a group of irreversible blinding diseases. Endogenous lipids may be involved in regulation of IOP homeostasis. We present comparative fold analysis of phospholipids and sphingolipids of aqueous humour and trabecular meshwork from human control vs primary open-angle glaucoma and mouse control (normotensive) vs ocular hypertensive state. The fold analysis in control vs disease state was based on ratiometric mass spectrometric data for above classes of lipids. We standardized in vitro assays for rapid characterization of lipids undergoing significant diminishment in disease state. Evaluation of lipids using in vitro assays helped select a finite number of lipids that may potentially expand cellular interstitial space embedded in an artificial matrix or increase fluid flow across a layer of cells. These assays reduced a number of lipids for initial evaluation using a mouse model, DBA/2J with spontaneous IOP elevation. These lipids were then used in other mouse models for confirmation of IOP lowering potential of a few lipids that were found promising in previous assessments. Our results provide selected lipid molecules that can be pursued for further evaluation and studies that may provide insight into their function.
Collapse
Affiliation(s)
- Genea Edwards
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami, Miami, FL, USA.,Department of Biochemistry and Molecular Biology, University of Miami, Miami, FL, USA
| | - Jennifer Arcuri
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami, Miami, FL, USA
| | - Haiyan Wang
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami, Miami, FL, USA.,Shanghai Key Laboratory of Ocular Fundus Diseases, Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Noel Ziebarth
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA
| | - Gulab Zode
- North Texas Eye Research Institute, University of North Texas, Fort Worth, TX, USA
| | - Richard K Lee
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami, Miami, FL, USA
| | - Sanjoy K Bhattacharya
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami, Miami, FL, USA.,Department of Biochemistry and Molecular Biology, University of Miami, Miami, FL, USA
| |
Collapse
|
14
|
Maddala R, Mongan M, Xia Y, Rao PV. Calponin-3 deficiency augments contractile activity, plasticity, fibrogenic response and Yap/Taz transcriptional activation in lens epithelial cells and explants. Sci Rep 2020; 10:1295. [PMID: 31992794 PMCID: PMC6987178 DOI: 10.1038/s41598-020-58189-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 01/09/2020] [Indexed: 01/19/2023] Open
Abstract
The transparent ocular lens plays a crucial role in vision by focusing light on to the retina with loss of lens transparency leading to impairment of vision. While maintenance of epithelial phenotype is recognized to be essential for lens development and function, knowledge of the identity of different molecular mechanisms regulating lens epithelial characteristics remains incomplete. This study reports that CNN-3, the acidic isoform of calponin, an actin binding contractile protein, is expressed preferentially and abundantly relative to the basic and neutral isoforms of calponin in the ocular lens, and distributes predominantly to the epithelium in both mouse and human lenses. Expression and MEKK1-mediated threonine 288 phosphorylation of CNN-3 is induced by extracellular cues including TGF-β2 and lysophosphatidic acid. Importantly, siRNA-induced deficiency of CNN3 in lens epithelial cell cultures and explants results in actin stress fiber reorganization, stimulation of focal adhesion formation, Yap activation, increases in the levels of α-smooth muscle actin, connective tissue growth factor and fibronectin, and decreases in E-cadherin expression. These results reveal that CNN3 plays a crucial role in regulating lens epithelial contractile activity and provide supporting evidence that CNN-3 deficiency is associated with the induction of epithelial plasticity, fibrogenic activity and mechanosensitive Yap/Taz transcriptional activation.
Collapse
Affiliation(s)
- Rupalatha Maddala
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Maureen Mongan
- Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Ying Xia
- Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Ponugoti Vasantha Rao
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, 27710, USA. .,Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, 27710, USA.
| |
Collapse
|
15
|
Ho LTY, Osterwald A, Ruf I, Hunziker D, Mattei P, Challa P, Vann R, Ullmer C, Rao PV. Role of the autotaxin-lysophosphatidic acid axis in glaucoma, aqueous humor drainage and fibrogenic activity. Biochim Biophys Acta Mol Basis Dis 2019; 1866:165560. [PMID: 31648019 DOI: 10.1016/j.bbadis.2019.165560] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 08/27/2019] [Accepted: 09/16/2019] [Indexed: 01/06/2023]
Abstract
Ocular hypertension due to impaired aqueous humor (AH) drainage through the trabecular meshwork (TM) is a major risk factor for glaucoma, a leading cause of irreversible blindness. However, the etiology of ocular hypertension remains unclear. Although autotaxin, a secreted lysophospholipase D and its catalytic product lysophosphatidic acid (LPA) have been shown to modulate AH drainage through TM, we do not have a complete understanding of their role and regulation in glaucoma patients, TM and AH outflow. This study reports a significant increase in the levels of autotaxin, lysophosphatidylcholine (LPC), LPA and connective tissue growth factor (CTGF) in the AH of Caucasian and African American open angle glaucoma patients relative to age-matched non-glaucoma patients. Treatment of human TM cells with dexamethasone, tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) increased the levels of autotaxin protein, a response that was mitigated by inhibitors of glucocorticoid receptor, NF-kB and SMAD3. Dexamethasone, TNF-α, IL-1β and LPC treatment of TM cells also led to an increase in the levels of CTGF, fibronectin and collagen type 1 in an autotaxin dependent manner. Additionally, in perfused enucleated mouse eyes, autotaxin and LPC were noted to decrease, while inhibition of autotaxin was increased aqueous outflow through the TM. Taken together, these results provide additional evidence for dysregulation of the autotaxin-LPA axis in the AH of glaucoma patients, reveal molecular insights into the regulation of autotaxin expression in TM cells and the consequences of autotaxin inhibitors in suppressing the fibrogenic response and resistance to AH outflow through the TM.
Collapse
Affiliation(s)
- Leona T Y Ho
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, USA
| | - Anja Osterwald
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Hoffmann-La Roche Ltd., Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Iris Ruf
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Hoffmann-La Roche Ltd., Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Daniel Hunziker
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Hoffmann-La Roche Ltd., Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Patrizio Mattei
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Hoffmann-La Roche Ltd., Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Pratap Challa
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, USA
| | - Robin Vann
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, USA
| | - Christoph Ullmer
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Hoffmann-La Roche Ltd., Grenzacherstrasse 124, 4070 Basel, Switzerland.
| | - Ponugoti Vasanth Rao
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, USA; Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
16
|
Rossi C, Cicalini I, Cufaro MC, Agnifili L, Mastropasqua L, Lanuti P, Marchisio M, De Laurenzi V, Del Boccio P, Pieragostino D. Multi-Omics Approach for Studying Tears in Treatment-Naïve Glaucoma Patients. Int J Mol Sci 2019; 20:ijms20164029. [PMID: 31426571 PMCID: PMC6721157 DOI: 10.3390/ijms20164029] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 08/13/2019] [Accepted: 08/14/2019] [Indexed: 12/20/2022] Open
Abstract
Primary open-angle glaucoma (POAG) represents the leading cause of irreversible blindness worldwide and is a multifactorial, chronic neurodegenerative disease characterized by retinal ganglion cell and visual field loss. There are many factors that are associated with the risk of developing POAG, with increased intraocular pressure being one of the most prevalent. Due to the asymptomatic nature of the disease, the diagnosis of POAG often occurs too late, which necessitates development of new effective screening strategies for early diagnosis of the disease. However, this task still remains unfulfilled. In order to provide further insights into the pathophysiology of POAG, we applied a targeted metabolomics strategy based on a high-throughput screening method for the determination of tear amino acids, free carnitine, acylcarnitines, succinylacetone, nucleosides, and lysophospholipids in naïve to therapy glaucomatous patients and normal controls. Also, we conducted proteomic analyses of the whole lacrimal fluid and purified extracellular vesicles obtained from POAG patients and healthy subjects. This multi-omics approach allowed us to conclude that POAG patients had lower levels of certain tear amino acids and lysophospholipids compared with controls. These targeted analyses also highlighted the low amount of acetylcarnitine (C2) in POAG patient which correlated well with proteomics data. Moreover, POAG tear proteins seemed to derive from extracellular vesicles, which carried a specific pro-inflammatory protein cargo.
Collapse
Affiliation(s)
- Claudia Rossi
- Center for Advanced Studies and Technology (CAST), University ''G. d'Annunzio'' of Chieti-Pescara, 66100 Chieti, Italy
- Department of Medical, Oral and Biotechnological Sciences, University ''G. d'Annunzio'' of Chieti-Pescara, 66100 Chieti, Italy
| | - Ilaria Cicalini
- Center for Advanced Studies and Technology (CAST), University ''G. d'Annunzio'' of Chieti-Pescara, 66100 Chieti, Italy
- Department of Medicine and Aging Science, "G. d'Annunzio" University of Chieti-Pescara, 66100 Chieti, Italy
| | - Maria Concetta Cufaro
- Center for Advanced Studies and Technology (CAST), University ''G. d'Annunzio'' of Chieti-Pescara, 66100 Chieti, Italy
- Department of Pharmacy, University ''G. d'Annunzio'' of Chieti-Pescara, 66100 Chieti, Italy
| | - Luca Agnifili
- Department of Medicine and Aging Science, "G. d'Annunzio" University of Chieti-Pescara, 66100 Chieti, Italy
- Opthalmic Clinic, Ss Annunziata Hospital, 66100 Chieti, Italy
| | - Leonardo Mastropasqua
- Department of Medicine and Aging Science, "G. d'Annunzio" University of Chieti-Pescara, 66100 Chieti, Italy
- Opthalmic Clinic, Ss Annunziata Hospital, 66100 Chieti, Italy
| | - Paola Lanuti
- Department of Medicine and Aging Science, "G. d'Annunzio" University of Chieti-Pescara, 66100 Chieti, Italy
| | - Marco Marchisio
- Department of Medicine and Aging Science, "G. d'Annunzio" University of Chieti-Pescara, 66100 Chieti, Italy
| | - Vincenzo De Laurenzi
- Center for Advanced Studies and Technology (CAST), University ''G. d'Annunzio'' of Chieti-Pescara, 66100 Chieti, Italy
- Department of Medical, Oral and Biotechnological Sciences, University ''G. d'Annunzio'' of Chieti-Pescara, 66100 Chieti, Italy
| | - Piero Del Boccio
- Center for Advanced Studies and Technology (CAST), University ''G. d'Annunzio'' of Chieti-Pescara, 66100 Chieti, Italy
- Department of Pharmacy, University ''G. d'Annunzio'' of Chieti-Pescara, 66100 Chieti, Italy
| | - Damiana Pieragostino
- Center for Advanced Studies and Technology (CAST), University ''G. d'Annunzio'' of Chieti-Pescara, 66100 Chieti, Italy.
- Department of Medical, Oral and Biotechnological Sciences, University ''G. d'Annunzio'' of Chieti-Pescara, 66100 Chieti, Italy.
| |
Collapse
|
17
|
Ho LTY, Skiba N, Ullmer C, Rao PV. Lysophosphatidic Acid Induces ECM Production via Activation of the Mechanosensitive YAP/TAZ Transcriptional Pathway in Trabecular Meshwork Cells. Invest Ophthalmol Vis Sci 2019; 59:1969-1984. [PMID: 29677358 PMCID: PMC5896423 DOI: 10.1167/iovs.17-23702] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Purpose Lysophosphatidic acid (LPA), a bioactive lipid, has been shown to increase resistance to aqueous humor outflow (AH) through the trabecular meshwork (TM). The molecular basis for this response of the TM to LPA, however, is not completely understood. In this study, we explored the possible involvement of mechanosensitive Yes-associated protein (YAP) and its paralog, transcriptional coactivator with PDZ-binding domain (TAZ), transcriptional activation in extracellular matrix (ECM) production by LPA-induced contractile activity in human TM cells (HTM). Methods The responsiveness of genes encoding LPA receptors (LPARs), LPA hydrolyzing lipid phosphate phosphatases (LPPs), and the LPA-generating autotaxin (ATX) to cyclic mechanical stretch in HTM cells, was evaluated by RT-quantitative (q)PCR. The effects of LPA and LPA receptor antagonists on actomyosin contractile activity, activation of YAP/TAZ, and levels of connective tissue growth factor (CTGF), and Cyr61 and ECM proteins in HTM cells were determined by immunoblotting, mass spectrometry, and immunofluorescence analyses. Results Cyclic mechanical stretch significantly increased the expression of several types of LPARs, LPP1, and ATX in HTM cells. LPA and LPA receptor–dependent contractile activity led to increases in both, the protein levels and activation of YAP/TAZ, and increased the levels of CTGF, Cyr61, α-smooth muscle actin (α–SMA), and ECM proteins in HTM cells. Conclusions The results of this study reveal that LPA and its receptors stimulate YAP/TAZ transcriptional activity in HTM cells by modulating cellular contractile tension, and augment expression of CTGF that in turn leads to increased production of ECM. Therefore, YAP/TAZ-induced increases in CTGF and ECM production could be an important molecular mechanism underlying LPA-induced resistance to AH outflow and ocular hypertension.
Collapse
Affiliation(s)
- Leona T Y Ho
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina, United States
| | - Nikolai Skiba
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina, United States
| | - Christoph Ullmer
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Ponugoti Vasantha Rao
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina, United States.,Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina, United States
| |
Collapse
|
18
|
Margolis MJ, Martinez M, Valencia J, Lee RK, Bhattacharya SK. Phospholipid secretions of organ cultured ciliary body. J Cell Biochem 2017; 119:2556-2566. [PMID: 28981155 DOI: 10.1002/jcb.26419] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 10/03/2017] [Indexed: 12/24/2022]
Abstract
Homeostasis of intraocular pressure (IOP) is important for the maintenance of anterior eye anatomic integrity, minimizing pressure-associated damage to the optic nerve, and maintaining a pressure gradient for blood flow to the eye. IOP is regulated by equilibrium between aqueous humor (AH) production and its outflow. The ciliary body (CB) is thought to actively secrete AH. However, whether AH composition and in particular, its phospholipids are entirely due to CB secretion remains uncertain. Comparison of phospholipids released by cultured CB, phospholipids present within CB tissue, within AH, and within blood and serum are consistent with release of most phospholipids into the AH by the CB. Treatment of CB in culture with timolol, a non-specific beta-adrenergic antagonist, alters the release of phospholipids by CB into the media. However, dorzalamide, a carbonic anhydrase inhibitor that reduces production of AH, does not affect phospholipid release thereby suggesting timolol, which also decreases IOP through decreased AH outflow, affects other physiological homeostatic mechanisms regulating aqueous outflow. These outflow changes also affect the composition of secreted phospholipids. We present evidence that release of lipids by the CB has a prolonged survival effect on cultured primary TM cells and TM tissue.
Collapse
Affiliation(s)
- Michael J Margolis
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Mitchell Martinez
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Jeffrey Valencia
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Richard K Lee
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Sanjoy K Bhattacharya
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida
| |
Collapse
|
19
|
Unravelling the interplay of sphingolipids and TGF-β signaling in the human corneal stroma. PLoS One 2017; 12:e0182390. [PMID: 28806736 PMCID: PMC5555661 DOI: 10.1371/journal.pone.0182390] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 07/17/2017] [Indexed: 11/19/2022] Open
Abstract
Purpose To delineate the role of Sphingolipids (SPLs) in the human cornea and their cross-talks with transforming growth factor beta (TGF-β) in order to develop novel, non-invasive therapies. Methods Human corneal fibroblasts (HCFs) were harvested from healthy donors, stimulated with Vitamin C to promote extracellular matrix assembly, treated with exogenous sphingosine-1-phosphate (S1P) or sphingosine kinase inhibitor 2 (SPHK I2) and isolated after 4 weeks for further analysis. Results Data showed that S1P led to a significant decrease in cellular migration where SPHK I2 just delayed it for 24h. Significant modulation of the sphingolipid pathway was also noted. Sphingosine kinase-1 (SphK1) was significantly downregulated upon exogenous stimulation with S1P at a concentration of 5μM and Sphingosine kinase-2 (SphK2) was also significantly downregulated at concentrations of 0.01μM, 0.1μM, and 5μM; whereas no effects were observed upon stimulation with SPHK I2. S1PR3 was significantly downregulated by 0.1μM and 5μM S1P and upregulated by 5μM and 10μM SPHK I2. Furthermore, both S1P and SPHK I2 regulated corneal fibrosis markers such as alpha-smooth muscle actin, collagen I, III, and V. We also investigated the interplay between two TGF-β isoforms and S1P/SPHK I2 treatments and found that TGF-β1 and TGF-β3 were both significantly upregulated with the 0.1μM S1P but were significantly downregulated with the 5μM S1P concentration. When TGF-β1 was compared directly to TGF-β3 expression, we observed that TGF-β3 was significantly downregulated compared to TGF-β1 in the 5μM concentration of S1P. No changes were observed upon SPHK I2 treatment. Conclusion Our study delineates the role of sphingolipids in the human cornea and highlights their different activities based on the cell/tissue type.
Collapse
|
20
|
|
21
|
Trabecular meshwork stiffness in glaucoma. Exp Eye Res 2016; 158:3-12. [PMID: 27448987 DOI: 10.1016/j.exer.2016.07.011] [Citation(s) in RCA: 121] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Revised: 07/15/2016] [Accepted: 07/18/2016] [Indexed: 12/26/2022]
Abstract
Alterations in stiffness of the trabecular meshwork (TM) may play an important role in primary open-angle glaucoma (POAG), the second leading cause of blindness. Specifically, certain data suggest an association between elevated intraocular pressure (IOP) and increased TM stiffness; however, the underlying link between TM stiffness and IOP remains unclear and requires further study. We here first review the literature on TM stiffness measurements, encompassing various species and based on a number of measurement techniques, including direct approaches such as atomic force microscopy (AFM) and uniaxial tension tests, and indirect methods based on a beam deflection model. We also briefly review the effects of several factors that affect TM stiffness, including lysophospholipids, rho-kinase inhibitors, cytoskeletal disrupting agents, dexamethasone (DEX), transforming growth factor-β2 (TGF-β2), nitric oxide (NO) and cellular senescence. We then describe a method we have developed for determining TM stiffness measurement in mice using a cryosection/AFM-based approach, and present preliminary data on TM stiffness in C57BL/6J and CBA/J mouse strains. Finally, we investigate the relationship between TM stiffness and outflow facility between these two strains. The method we have developed shows promise for further direct measurements of mouse TM stiffness, which may be of value in understanding mechanistic relations between outflow facility and TM biomechanical properties.
Collapse
|
22
|
Lerner N, Beit-Yannai E. Cross-talk between ciliary epithelium and trabecular meshwork cells in-vitro: a new insight into glaucoma. PLoS One 2014; 9:e112259. [PMID: 25389776 PMCID: PMC4229184 DOI: 10.1371/journal.pone.0112259] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 10/06/2014] [Indexed: 12/17/2022] Open
Abstract
PURPOSE It is assumed that the non-pigmented ciliary epithelium plays a role in regulating intraocular pressure via its neuroendocrine activities. To test this hypothesis, we investigated the effect on a human trabecular meshwork (TM) cell line (NTM) of co-culture with a human non-pigmented ciliary epithelium cell line (ODM-2). METHODS The cellular cross-talk between ODM-2 and NTM cells was studied in a co-culture system in which the two cell types were co-cultured for 5 to 60 min or 2, 4 and 8h and then removed from the co-culture and analyzed. Analyses of the ERK and p38 mitogen-activated protein kinase (MAPK) pathways and of the activity of TM phosphatases and matrix metalloproteins (MMPs) were performed. Acid and alkaline phosphatase activity was determined by the DiFMUP (6, 8-difluoro-4-methylumbelliferyl phosphate) assay. MMP levels were determined by gelatin zymography. RESULTS Exposure of NTM cells to ODM-2 cells led to the activation of the MAPK signal transduction pathways in NTM cells within 5 min of co-culture. Phosphorylation of ERK1/ERK2 and p38 peaked at 10 and 15 min and then decreased over time. Interaction between ODM-2 and NTM cells promoted the expression of MMP-9 in the NTM cells after 4h of co-culture. CONCLUSIONS Our findings provide support for the hypothesis that crosstalk does indeed take place between ODM-2 and NTM cells. Future studies should be designed to determine the relationship between the MMP system, MAPK kinases and phosphatases. Manipulation of these signaling molecules and the related NTM signal transduction pathways may provide targets for developing improved treatments for glaucoma.
Collapse
Affiliation(s)
- Natalie Lerner
- Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Elie Beit-Yannai
- Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| |
Collapse
|
23
|
Edwards G, Aribindi K, Guerra Y, Bhattacharya SK. Sphingolipids and ceramides of mouse aqueous humor: Comparative profiles from normotensive and hypertensive DBA/2J mice. Biochimie 2014; 105:99-109. [PMID: 25014247 DOI: 10.1016/j.biochi.2014.06.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Accepted: 06/25/2014] [Indexed: 11/17/2022]
Abstract
PURPOSE To identify the sphingolipid and ceramide species and their quantitative differences between normotensive and hypertensive intraocular pressure states in DBA/2J mouse aqueous humor (AH). METHODS Normotensive and hypertensive AH was sampled from mice by paracentesis. Lipid extraction was performed using modifications of the Bligh and Dyer method. Protein concentration was estimated using the Bradford colorimetric assay. Sphingolipids and ceramides were identified and subjected to ratiometric quantification using appropriate class specific lipid standards on a TSQ Quantum Access Max triple quadrupole mass spectrometer. RESULTS The comparative profiles of normotensive and hypertensive DBA/2J mouse AH showed several species of sphingomyelin, sphingoid base, sphingoid base-1-phosphate (S1P) and ceramides common between them. A number of unique lipids in each of the above lipid classes were also identified in normotensive AH that were absent in hypertensive AH and vice versa. CONCLUSION A number of sphingolipid and ceramide species were found to be uniquely present in normotensive, but absent in hypertensive AH and vice versa. Further pursuit of these findings is likely to contribute towards expanding our understanding of the molecular changes associated with increased intraocular pressure (IOP) and glaucoma.
Collapse
Affiliation(s)
- Genea Edwards
- Bascom Palmer Eye Institute, University of Miami School of Medicine, Miami, FL 33136, USA
| | - Katyayini Aribindi
- Bascom Palmer Eye Institute, University of Miami School of Medicine, Miami, FL 33136, USA
| | - Yenifer Guerra
- Bascom Palmer Eye Institute, University of Miami School of Medicine, Miami, FL 33136, USA
| | - Sanjoy K Bhattacharya
- Bascom Palmer Eye Institute, University of Miami School of Medicine, Miami, FL 33136, USA.
| |
Collapse
|