1
|
Sumer F, Subasi S, Bahceci I, Satilmaz F. Evaluation of serum galectin-3 concentration as a potential biomarker in exudative-type age-related macular degeneration. Sci Rep 2024; 14:31957. [PMID: 39738409 DOI: 10.1038/s41598-024-83499-w] [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: 08/28/2024] [Accepted: 12/16/2024] [Indexed: 01/02/2025] Open
Abstract
To investigate the effect of serum galectin-3 on naive neovascular AMD and its use as a serum marker by revealing the variation in this molecule between patient and control groups. Fifty-six naive neovascular AMD patients and 30 healthy control age-matched healthy subjects were included in this prospective case‒control study. Blood samples were obtained and used for analysis of complete blood count; High sensitivity C-reactive protein (HsCRP), erythrocyte sedimentation rate (ESR), low-density lipoprotein (LDL), high-density lipoprotein (HDL), triglyceride (TG), total cholesterol, homocysteine, HbA1c and galectin-3 levels. The average HsCRP level in the AMD group was significantly higher than that in the control group (p < 0.001). The median leukocyte count was significantly higher in the AMD group than in the control group (p < 0.001). Total cholesterol, LDL and TG levels were significantly higher in the AMD group than in the control group (p < 0.001; in all comparisons). The mean HDL level was significantly lower in the AMD group than in the control group (p < 0.001). The mean galectin-3 level was 8.79 ± 0.55 in the AMD group and 6.55 ± 0.55 in the control group. There was a statistically significant increase in galectin-3 levels in the AMD group (p < 0.001). There was a significant positive correlation between CMT and galectin-3 levels in the control (r = 0.495, p = 0.005) and AMD (r = 0.776, p < 0.001) groups. This study reports that serum Gal-3 concentration was higher in the AMD group compared to the control group and positively correlated with CMT.
Collapse
Affiliation(s)
- Fatma Sumer
- Department of Ophthalmology, Faculty of Medicine, Recep Tayyip Erdogan University, Rize, 53100, Turkey.
| | - Sevgi Subasi
- Department of Ophthalmology, Faculty of Medicine, Kocaeli University, Kocaeli, 41100, Turkey
| | - Ilkay Bahceci
- Department of Microbiology, Faculty of Medicine, Recep Tayyip Erdogan University, Rize, 53100, Turkey
| | - Fatih Satilmaz
- Department of Ophthalmology, Faculty of Medicine, Recep Tayyip Erdogan University, Rize, 53100, Turkey
| |
Collapse
|
2
|
Stanić R, Vukojević K, Filipović N, Benzon B, Ogorevc M, Kunac N, Čanović S, Kovačević P, Paradžik Šimunović M, Konjevoda S. The Effect of Prostaglandin F2 Analog Treatment on the Immunoexpression of Fibrosis-Associated Factors in Patients with Glaucoma Undergoing Deep Sclerotomy. Int J Mol Sci 2024; 25:12618. [PMID: 39684329 DOI: 10.3390/ijms252312618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 11/17/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Long-term use of topical prostaglandins might initiate chronic conjunctival inflammation, leading to poor outcomes of glaucoma surgery. The aim of this study was to evaluate the immunoexpression pattern of HSP70, CTGF, SNAIL, aSMA, cMYB, and HIFa in the conjunctiva, episclera, and deep sclera in patients with glaucoma undergoing deep sclerectomy in order to establish an association between staining intensities and prostaglandin F2 (PGF2) treatment. Double immunofluorescence (HSP70, CTGF, SNAIL, aSMA, cMYB, and HIFa) was performed on conjunctiva, episclera, and deep sclera samples, which were obtained from 23 patients treated with PGF2 and 8 patients without PGF2 treatment. When comparing the ocular tissues of patients regarding treatment with PGF2 analogs, we found a significant increase in the immunoexpression of HSP70 in the conjunctival epithelium of patients treated with PGF2 analogs compared to those without PGF2 treatment. These patients also had an increase in SNAIL immunoexpression and a decrease in aSMA immunoexpression in the deep sclera. There were no significant differences in HIFa, CTGF, or cMYB immunoexpression levels between the two groups. Further research into the regulation of these factors in ocular tissues could lead to the development of potential novel therapeutic approaches in glaucoma management.
Collapse
Affiliation(s)
- Robert Stanić
- Department of Ophthalmology, University Hospital in Split, Šoltanska 1, 21000 Split, Croatia
| | - Katarina Vukojević
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Šoltanska 2, 21000 Split, Croatia
| | - Natalija Filipović
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Šoltanska 2, 21000 Split, Croatia
| | - Benjamin Benzon
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Šoltanska 2, 21000 Split, Croatia
| | - Marin Ogorevc
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Šoltanska 2, 21000 Split, Croatia
| | - Nenad Kunac
- Department of Pathology, Forensic Medicine and Cytology, University Hospital in Split, Spinčićeva 1, 21000 Split, Croatia
| | - Samir Čanović
- Department of Ophtalmology, General Hospital Zadar, Ul. Bože Peričića 5, 23000 Zadar, Croatia
| | - Petra Kovačević
- Department of Ophthalmology, University Hospital Centre Zagreb, Kišpatićeva 12, 10000 Zagreb, Croatia
| | | | - Suzana Konjevoda
- Department of Ophtalmology, General Hospital Zadar, Ul. Bože Peričića 5, 23000 Zadar, Croatia
- Department of Health Studies, University of Zadar, Ulica Mihovila Pavlinovica 1, 23000 Zadar, Croatia
| |
Collapse
|
3
|
Salkar A, Wall RV, Basavarajappa D, Chitranshi N, Parilla GE, Mirzaei M, Yan P, Graham S, You Y. Glial Cell Activation and Immune Responses in Glaucoma: A Systematic Review of Human Postmortem Studies of the Retina and Optic Nerve. Aging Dis 2024; 15:2069-2083. [PMID: 38502591 PMCID: PMC11346413 DOI: 10.14336/ad.2024.0103] [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: 10/30/2023] [Accepted: 01/03/2024] [Indexed: 03/21/2024] Open
Abstract
Although researched extensively the understanding regarding mechanisms underlying glaucoma pathogenesis remains limited. Further, the exact mechanism behind neuronal death remains elusive. The role of neuroinflammation in retinal ganglion cell (RGC) death has been prominently theorised. This review provides a comprehensive summary of neuroinflammatory responses in glaucoma. A systematic search of Medline and Embase for articles published up to 8th March 2023 yielded 32 studies using post-mortem tissues from glaucoma patients. The raw data were extracted from tables and text to calculate the standardized mean differences (SMDs). These studies utilized post-mortem tissues from glaucoma patients, totalling 490 samples, compared with 380 control samples. Among the included studies, 27 reported glial cell activation based on changes to cellular morphology and molecular staining. Molecular changes were predominantly attributed to astrocytes (62.5%) and microglia (15.6%), with some involvement of Muller cells. These glial cell changes included amoeboid microglial cells with increased CD45 or HLA-DR intensity and hypertrophied astrocytes with increased glial fibrillary acidic protein labelling. Further, changes to extracellular matrix proteins like collagen, galectin, and tenascin-C suggested glial cells' influence on structural changes in the optic nerve head. The activation of DAMPs-driven immune response and the classical complement cascade was reported and found to be associated with activated glial cells in glaucomatous tissue. Increased pro-inflammatory markers such as interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-α) were also linked to glial cells. Glial cell activation was also associated with mitochondrial, vascular, metabolic and antioxidant component disruptions. Association of the activated glial cells with pro-inflammatory responses, dysregulation of homeostatic components and antigen presentation indicates that glial cell responses influence glaucoma progression. However, the exact mechanism triggering these responses and underlying interactions remains unexplored. This necessitates further research using human samples for an increased understanding of the precise role of neuroinflammation in glaucoma progression.
Collapse
Affiliation(s)
- Akanksha Salkar
- Department of Clinical Medicine, Faculty of Human, Health, and Medical Science, Macquarie University. Sydney, NSW, Australia.
| | - Roshana Vander Wall
- Department of Clinical Medicine, Faculty of Human, Health, and Medical Science, Macquarie University. Sydney, NSW, Australia.
| | - Devaraj Basavarajappa
- Department of Clinical Medicine, Faculty of Human, Health, and Medical Science, Macquarie University. Sydney, NSW, Australia.
| | - Nitin Chitranshi
- Department of Clinical Medicine, Faculty of Human, Health, and Medical Science, Macquarie University. Sydney, NSW, Australia.
| | - Gabriella E. Parilla
- Department of Clinical Medicine, Faculty of Human, Health, and Medical Science, Macquarie University. Sydney, NSW, Australia.
| | - Mehdi Mirzaei
- Department of Clinical Medicine, Faculty of Human, Health, and Medical Science, Macquarie University. Sydney, NSW, Australia.
| | - Peng Yan
- Department of Ophthalmology & Vision Sciences, University of Toronto, Kensington Eye Institute/UHN, Canada.
| | - Stuart Graham
- Department of Clinical Medicine, Faculty of Human, Health, and Medical Science, Macquarie University. Sydney, NSW, Australia.
| | - Yuyi You
- Department of Clinical Medicine, Faculty of Human, Health, and Medical Science, Macquarie University. Sydney, NSW, Australia.
- Save Sight Institute, University of Sydney. Sydney, NSW, Australia.
| |
Collapse
|
4
|
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
|
5
|
Zhou Z, Feng Z, Sun X, Wang Y, Dou G. The Role of Galectin-3 in Retinal Degeneration and Other Ocular Diseases: A Potential Novel Biomarker and Therapeutic Target. Int J Mol Sci 2023; 24:15516. [PMID: 37958500 PMCID: PMC10649114 DOI: 10.3390/ijms242115516] [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: 10/09/2023] [Revised: 10/17/2023] [Accepted: 10/19/2023] [Indexed: 11/15/2023] Open
Abstract
Galectin-3 is the most studied member of the Galectin family, with a large range of mediation in biological activities such as cell growth, proliferation, apoptosis, differentiation, cell adhesion, and tissue repair, as well as in pathological processes such as inflammation, tissue fibrosis, and angiogenesis. As is known to all, inflammation, aberrant cell apoptosis, and neovascularization are the main pathophysiological processes in retinal degeneration and many ocular diseases. Therefore, the review aims to conclude the role of Gal3 in the retinal degeneration of various diseases as well as the occurrence and development of the diseases and discuss its molecular mechanisms according to research in systemic diseases. At the same time, we summarized the predictive role of Gal3 as a biomarker and the clinical application of its inhibitors to discuss the possibility of Gal3 as a novel target for the treatment of ocular diseases.
Collapse
Affiliation(s)
| | | | | | - Yusheng Wang
- Department of Ophthalmology, Eye Institute of Chinese PLA, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China; (Z.Z.); (Z.F.); (X.S.)
| | - Guorui Dou
- Department of Ophthalmology, Eye Institute of Chinese PLA, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China; (Z.Z.); (Z.F.); (X.S.)
| |
Collapse
|
6
|
Cullen PF, Sun D. Astrocytes of the eye and optic nerve: heterogeneous populations with unique functions mediate axonal resilience and vulnerability to glaucoma. FRONTIERS IN OPHTHALMOLOGY 2023; 3:1217137. [PMID: 37829657 PMCID: PMC10569075 DOI: 10.3389/fopht.2023.1217137] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
Abstract
The role of glia, particularly astrocytes, in mediating the central nervous system's response to injury and neurodegenerative disease is an increasingly well studied topic. These cells perform myriad support functions under physiological conditions but undergo behavioral changes - collectively referred to as 'reactivity' - in response to the disruption of neuronal homeostasis from insults, including glaucoma. However, much remains unknown about how reactivity alters disease progression - both beneficially and detrimentally - and whether these changes can be therapeutically modulated to improve outcomes. Historically, the heterogeneity of astrocyte behavior has been insufficiently addressed under both physiological and pathological conditions, resulting in a fragmented and often contradictory understanding of their contributions to health and disease. Thanks to increased focus in recent years, we now know this heterogeneity encompasses both intrinsic variation in physiological function and insult-specific changes that vary between pathologies. Although previous studies demonstrate astrocytic alterations in glaucoma, both in human disease and animal models, generally these findings do not conclusively link astrocytes to causative roles in neuroprotection or degeneration, rather than a subsequent response. Efforts to bolster our understanding by drawing on knowledge of brain astrocytes has been constrained by the primacy in the literature of findings from peri-synaptic 'gray matter' astrocytes, whereas much early degeneration in glaucoma occurs in axonal regions populated by fibrous 'white matter' astrocytes. However, by focusing on findings from astrocytes of the anterior visual pathway - those of the retina, unmyelinated optic nerve head, and myelinated optic nerve regions - we aim to highlight aspects of their behavior that may contribute to axonal vulnerability and glaucoma progression, including roles in mitochondrial turnover and energy provisioning. Furthermore, we posit that astrocytes of the retina, optic nerve head and myelinated optic nerve, although sharing developmental origins and linked by a network of gap junctions, may be best understood as distinct populations residing in markedly different niches with accompanying functional specializations. A closer investigation of their behavioral repertoires may elucidate not only their role in glaucoma, but also mechanisms to induce protective behaviors that can impede the progressive axonal damage and retinal ganglion cell death that drive vision loss in this devastating condition.
Collapse
Affiliation(s)
- Paul F. Cullen
- Department of Ophthalmology, Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, United States
| | - Daniel Sun
- Department of Ophthalmology, Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
7
|
Palchunova K, Kaji Y, Fujita A, Oshika T. RNA-seq analysis of human trabecular endothelial cells after treatment with timolol maleate. Jpn J Ophthalmol 2023:10.1007/s10384-023-00998-5. [PMID: 37314597 DOI: 10.1007/s10384-023-00998-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 04/26/2023] [Indexed: 06/15/2023]
Abstract
PURPOSE Timolol maleate (timolol), a β-receptor blocker, reduces intraocular pressure by decreasing aqueous humor production. Timolol reportedly also protects ganglion cells, decreases aqueous humor outflow facility, and destroys the extracellular matrix in the trabecular meshwork. In this study, we investigated the effects of timolol on cultured human trabecular endothelial cells purchased from ScienCell using next-generation sequencing. STUDY DESIGN Experimental investigation. METHODS Total ribonucleic acid (RNA) was extracted after 24 h. More than 100 million RNAs in control and timolol-treated group were sequenced using a next-generation sequencer. The expression of 55,778 RNAs was analyzed. RESULTS A total of 2,105 genes were significantly upregulated and 2,125 genes were downregulated, after the addition of timolol. VGF nerve growth factor inducible (VGF) (388-fold) had the maximum increase in expression, followed by amphiregulin (333-fold), a member of the epidermal growth factor family. Moreover, the expression of extracellular matrix-degrading enzymes, matrix metalloproteinases (MMPs) 1, 2, 3, 10, 12, and 14, increased. CONCLUSION Timolol exerts various effects on human trabecular endothelial cells. The increase in MMP expression may contribute to the decrease in the aqueous humor outflow facility.
Collapse
Affiliation(s)
- Kseniya Palchunova
- Department of Ophthalmology, University of Tsukuba Majors in Medical Sciences, 1-1-1 Tennoudai Tsukuba, Ibaraki, 3058575, Japan
| | - Yuichi Kaji
- Department of Ophthalmology, University of Tsukuba Majors in Medical Sciences, 1-1-1 Tennoudai Tsukuba, Ibaraki, 3058575, Japan.
- Matsumoto Eye Clinic, 2-25-2F Toride-i-center, Chuo-machi Toride, Ibaraki, 3020014, Japan.
| | - Akari Fujita
- Kagurazaka Eye Clinic, 115 Yarai-cho Shinjyuku, Tokyo, 1620805, Japan
| | - Tetsuro Oshika
- Department of Ophthalmology, University of Tsukuba Majors in Medical Sciences, 1-1-1 Tennoudai Tsukuba, Ibaraki, 3058575, Japan
| |
Collapse
|
8
|
Rangan R, Sad do Valle R, Tovar-Vidales T. Expression of procollagen C-proteinase enhancer 1 in human trabecular meshwork tissues and cells. Exp Eye Res 2022; 225:109280. [PMID: 36252654 DOI: 10.1016/j.exer.2022.109280] [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: 04/25/2022] [Revised: 10/03/2022] [Accepted: 10/08/2022] [Indexed: 12/29/2022]
Abstract
Glaucoma is a primary cause of progressive, irreversible blindness. One of the primary tissues involved in glaucoma pathology is the trabecular meshwork (TM). In glaucoma, the TM is a site of increased extracellular matrix (ECM) protein secretion, deposition, and accumulation, contributing to a disrupted TM architecture and increased resistance to the outflow of aqueous humor. The healthy TM structure is comprised of sheets and beams composed of multiple extracellular matrix proteins, including mature fibrillar collagens. In the glaucomatous eye, this structure is disrupted by the abnormal deposition of collagen fibrils and other ECM proteins in the TM. In this study, we determined whether procollagen C-proteinase enhancer 1 (PCOLCE1) - a protein typically involved in collagen fibril processing - is expressed in the human TM tissues and cells and whether its expression is altered in glaucomatous conditions. Using immunocytochemistry, qPCR, and western blot (WB) analyses, we found that PCOLCE1 is expressed and translated in human TM tissues and cells. Our data analysis suggests that PCOLCE1 expression by TM cells may be downregulated by TGFβ2 treatment, which warrants further investigation of a possible role for PCOLCE1 in glaucomatous pathology.
Collapse
Affiliation(s)
- Rajiv Rangan
- 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, 76107-2699, USA
| | - Rafael Sad do Valle
- 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, 76107-2699, USA
| | - Tara Tovar-Vidales
- 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, 76107-2699, USA.
| |
Collapse
|
9
|
Pitts KM, Neeson CE, Hall NE, Lin JB, Falah HK, Wang SL, Lo KT, Song CE, Margeta MA, Solá-Del Valle DA. Neurodegeneration Markers Galectin-3 and Apolipoprotein E Are Elevated in the Aqueous Humor of Eyes With Glaucoma. Transl Vis Sci Technol 2022; 11:1. [DOI: 10.1167/tvst.11.11.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Affiliation(s)
- Kristen M. Pitts
- Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Cameron E. Neeson
- Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Nathan E. Hall
- Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Jonathan B. Lin
- Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Henisk K. Falah
- Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Silas L. Wang
- Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Kristine T. Lo
- Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Christian E. Song
- Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Milica A. Margeta
- Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - David A. Solá-Del Valle
- Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
10
|
Animal Model Contributions to Primary Congenital Glaucoma. J Ophthalmol 2022; 2022:6955461. [PMID: 35663518 PMCID: PMC9162845 DOI: 10.1155/2022/6955461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 05/12/2022] [Indexed: 11/17/2022] Open
Abstract
Primary congenital glaucoma (PCG) is an ocular disease characterized by congenital anterior segmental maldevelopment with progressive optic nerve degeneration. Certain genes, such as cytochrome P450 family 1 subfamily B member 1 and latent TGF-β-binding protein 2, are involved in the pathogenesis of PCG, but the exact pathogenic mechanism has not yet been fully elucidated. There is an urgent need to determine the etiology and pathophysiology of PCG and develop new therapeutic methods to stop disease progression. Animal models can simulate PCG and are essential to study the pathogenesis and treatment of PCG. Various animal species have been used in the study of PCG, including rabbits, rats, mice, cats, zebrafish, and quails. These models are formed spontaneously or by combining with genetic engineering technology. The focus of the present study is to review the characteristics and potential applications of animal models in PCG and provide new approaches to understand the mechanism and develop new treatment strategies for patients with PCG.
Collapse
|
11
|
Pumphrey SA, Zitek-Morrison E, Pizzirani S, Meola DM. Evaluation of matrix metalloproteinases and tissue inhibitors of metalloproteinases in aqueous humor of dogs with versus without naturally occurring primary angle-closure glaucoma. Am J Vet Res 2021; 83:245-255. [PMID: 34936570 DOI: 10.2460/ajvr.21.04.0062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To compare concentrations of matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinases (TIMPs) in aqueous humor from ophthalmologically normal dogs and dogs with naturally occurring primary angle-closure glaucoma (cPACG). SAMPLE Aqueous humor samples from 12 eyes with cPACG and 18 ophthalmologically normal eyes of dogs. PROCEDURES A multiplex fluorescence-based ELISA was used to measure concentrations of MMP-1, MMP-2, MMP-3, MMP-8, MMP-9, MMP-10, MMP-13, TIMP-1, TIMP-2, and TIMP-4. Results for eyes with versus without cPACG were compared. RESULTS Significantly higher mean concentrations of MMP-1 (45% higher), MMP-2 (55% higher), MMP-3 (39% higher), MMP-8 (79% higher), MMP-9 (29% higher), MMP-10 (60% higher), TIMP-1 (63% higher), and TIMP-2 (136% higher) were detected in aqueous humor from eyes with cPACG, compared with ophthalmologically normal eyes. CLINICAL RELEVANCE MMPs and TIMPs have pivotal roles in extracellular matrix turnover and homeostasis in the outflow pathways of the eye. Results of the present study documented higher concentrations of MMPs and TIMPs in aqueous humor samples from dog eyes with late-stage cPACG. Although, to our knowledge, TIMPs have not previously been evaluated in the context of cPACG, the markedly higher concentration of TIMPs in eyes with cPACG suggested that inhibition of proteolysis and extracellular matrix turnover might be a factor in the development of glaucoma in susceptible individuals. However, because the present study used samples from dogs with late-stage cPACG, further work is required to characterize the temporal relationship between MMP and TIMP concentration changes and onset or progression of disease.
Collapse
Affiliation(s)
- Stephanie A Pumphrey
- Department of Clinical Sciences, Cummings School of Veterinary Medicine, Tufts University, North Grafton, MA
| | - Emily Zitek-Morrison
- Department of Population and Quantitative Health Sciences, UMass Chan Medical School, University of Massachusetts, Worcester, MA
| | - Stefano Pizzirani
- Department of Clinical Sciences, Cummings School of Veterinary Medicine, Tufts University, North Grafton, MA
| | - Dawn M Meola
- Department of Clinical Sciences, Cummings School of Veterinary Medicine, Tufts University, North Grafton, MA
| |
Collapse
|
12
|
Rezaei Kanavi M, Yazdani S, Elahi E, Mirrahimi M, Hajizadeh M, Khodaverdi S, Suri F. Prenatal diagnosis of primary congenital glaucoma and histopathological features in a fetal globe with cytochrome p4501B1 mutations. Eur J Ophthalmol 2021; 32:11206721211051235. [PMID: 34730456 DOI: 10.1177/11206721211051235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND/OBJECTIVES This study aims to report the developmental and histopathological features of ocular tissues from an electively aborted human fetus with mutations in cytochrome p4501B1, and thus predisposed to primary congenital glaucoma in comparison to an age-matched healthy fetal globe. SUBJECTS/METHODS Both eyes of two 17-week gestational aged fetuses, the first with CYP1B1 mutations and the second as healthy control fetus, were studied. Hematoxylin and eosin, Periodic acid-Schiff, Gomori's trichrome, and Verhoeff-Van Gieson staining protocols in addition to immunohistochemistry staining using anti-cytochrome p4501B1, anti-fibrillin-1, and anti-4-hydroxy-2-nonenal antibodies, as primary antibodies, were performed to assess the effect of the mutations on tissue development, cytochrome p4501B1 protein expression, extracellular matrix structure, and oxidative stress in the developing fetus eye. Quantitative analyses were performed using ImageJ software. Student's t-test was used for statistical analysis and P-values <0.05 were considered as significant. RESULTS Delayed development in ocular tissues, decreased expression of cytochrome p4501B1 protein, irregular extracellular matrix structure, and increased oxidative stress biomarker were evident in the ocular tissues of the fetus with cytochrome p4501B1 mutations as compared to a normal globe from an age-matched fetus. CONCLUSION To the best of our knowledge, this is the first report of prenatal diagnosis of primary congenital glaucoma. We also describe histopathological changes in the primary congenital glaucoma-affected globes revealing the effect of cytochrome p4501B1 deficiency on ocular tissues during early fetal development contributing to the glaucoma phenotype.
Collapse
Affiliation(s)
- Mozhgan Rezaei Kanavi
- Ocular Tissue Engineering Research Center, Research Institute for Ophthalmology and Vision Science, 556492Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shahin Yazdani
- Ocular Tissue Engineering Research Center, Research Institute for Ophthalmology and Vision Science, 556492Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Elahe Elahi
- School of Biology, University College of Science, 48425University of Tehran, Tehran, Iran
| | - Mehraban Mirrahimi
- Ophthalmic Research Center, Research Institute for Ophthalmology and Vision Science, 556492Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Hajizadeh
- Ophthalmic Research Center, Research Institute for Ophthalmology and Vision Science, 556492Shahid Beheshti University of Medical Sciences, Tehran, Iran
- 226735Department of Ophthalmology, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Sepideh Khodaverdi
- Department of Obstetrics and Gynecology, Endometriosis Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Suri
- Ophthalmic Research Center, Research Institute for Ophthalmology and Vision Science, 556492Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
13
|
Mirna Expression in Glaucomatous and TGFβ2 Treated Lamina Cribrosa Cells. Int J Mol Sci 2021; 22:ijms22126178. [PMID: 34201109 PMCID: PMC8229860 DOI: 10.3390/ijms22126178] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/27/2021] [Accepted: 06/04/2021] [Indexed: 01/25/2023] Open
Abstract
Glaucoma is a group of optic neuropathies that leads to irreversible vision loss. The optic nerve head (ONH) is the site of initial optic nerve damage in glaucoma. ONH-derived lamina cribrosa (LC) cells synthesize extracellular matrix (ECM) proteins; however, these cells are adversely affected in glaucoma and cause detrimental changes to the ONH. LC cells respond to mechanical strain by increasing the profibrotic cytokine transforming growth factor-beta 2 (TGFβ2) and ECM proteins. Moreover, microRNAs (miRNAs or miR) regulate ECM gene expression in different fibrotic diseases, including glaucoma. A delicate homeostatic balance between profibrotic and anti-fibrotic miRNAs may contribute to the remodeling of ONH. This study aimed to determine whether modulation of miRNAs alters the expression of ECM in human LC cells. Primary human normal and glaucoma LC cells were grown to confluency and treated with or without TGFβ2 for 24 h. Differences in expression of miRNAs were analyzed using miRNA qPCR arrays. miRNA PCR arrays showed that the miR-29 family was significantly decreased in glaucomatous LC cell strains compared to age-matched controls. TGFβ2 treatment downregulated the expression of multiple miRNAs, including miR-29c-3p, compared to controls in LC cells. LC cells transfected with miR-29c-3p mimics or inhibitors modulated collagen expression.
Collapse
|
14
|
Wan P, Long E, Li Z, Zhu Y, Su W, Zhuo Y. TET-dependent GDF7 hypomethylation impairs aqueous humor outflow and serves as a potential therapeutic target in glaucoma. Mol Ther 2021; 29:1639-1657. [PMID: 33388417 PMCID: PMC8058441 DOI: 10.1016/j.ymthe.2020.12.030] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 10/15/2020] [Accepted: 12/18/2020] [Indexed: 01/03/2023] Open
Abstract
Glaucoma is the leading cause of irreversible vision loss, affecting more than 70 million individuals worldwide. Circulatory disturbances of aqueous humor (AH) have long been central pathological contributors to glaucomatous lesions. Thus, targeting the AH outflow is a promising approach to treat glaucoma. However, the epigenetic mechanisms initiating AH outflow disorders and the targeted treatments remain to be developed. Studying glaucoma patients, we identified GDF7 (growth differentiation factor 7) hypomethylation as a crucial event in the onset of AH outflow disorders. Regarding the underlying mechanism, the hypomethylated GDF7 promoter was responsible for the increased GDF7 production and secretion in primary open-angle glaucoma (POAG). Excessive GDF7 protein promoted trabecular meshwork (TM) fibrosis through bone morphogenetic protein receptor type 2 (BMPR2)/Smad signaling and upregulated pro-fibrotic genes, α-smooth muscle actin (α-SMA) and fibronectin (FN). GDF7 protein expression formed a positive feedback loop in glaucomatous TM (GTM). This positive feedback loop was dependent on the activated TET (ten-eleven translocation) enzyme, which kept the GDF7 promoter region hypomethylated. The phenotypic transition in TM fortified the AH outflow resistance, thus elevating the intraocular pressure (IOP) and attenuating the nerve fiber layer. This methylation-dependent mechanism is also confirmed by a machine-learning model in silico with a specificity of 84.38% and a sensitivity of 89.38%. In rhesus monkeys, we developed GDF7 neutralization therapy to inhibit TM fibrosis and consequent AH outflow resistance that contributes to glaucoma. The neutralization therapy achieved high-efficiency control of the IOP (from 21.3 ± 0.3 to 17.6 ± 0.2 mmHg), a three-fold improvement in the outflow facility (from 0.1 to 0.3 μL/min · mmHg), and protection of nerve fibers. This study provides new insights into the epigenetic mechanism of glaucoma and proposes an innovative GDF7 neutralization therapy as a promising intervention.
Collapse
Affiliation(s)
- Peixing Wan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China; Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Erping Long
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China; Department of Ecology and Evolutionary Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Zhidong Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Yingting Zhu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Wenru Su
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Yehong Zhuo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China.
| |
Collapse
|
15
|
Tribble JR, Kokkali E, Otmani A, Plastino F, Lardner E, Vohra R, Kolko M, André H, Morgan JE, Williams PA. When Is a Control Not a Control? Reactive Microglia Occur Throughout the Control Contralateral Pathway of Retinal Ganglion Cell Projections in Experimental Glaucoma. Transl Vis Sci Technol 2021; 10:22. [PMID: 33510961 PMCID: PMC7804521 DOI: 10.1167/tvst.10.1.22] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 12/02/2020] [Indexed: 12/19/2022] Open
Abstract
Purpose Animal models show retinal ganglion cell (RGC) injuries that replicate features of glaucoma and the contralateral eye is commonly used as an internal control. There is significant crossover of RGC axons from the ipsilateral to the contralateral side at the level of the optic chiasm, which may confound findings when damage is restricted to one eye. The effect of unilateral glaucoma on neuroinflammatory damage to the contralateral pathway of RGC projections has largely been unexplored. Methods Ocular hypertensive glaucoma was induced unilaterally or bilaterally in the rat and RGC neurodegenerative events were assessed. Neuroinflammation was quantified in the retina, optic nerve head, optic nerve, lateral geniculate nucleus, and superior colliculus by high-resolution imaging, and in the retina by flow cytometry and protein arrays. Results After ocular hypertensive stress, peripheral monocytes enter the retina and microglia become reactive. This effect is more marked in animals with bilateral ocular hypertensive glaucoma. In rats where glaucoma was induced unilaterally, there was significant microglia activation in the contralateral (control) eye. Microglial activation extended into the optic nerve and terminal visual thalami, where it was similar across hemispheres in unilateral ocular hypertension. Conclusions These data suggest that caution is warranted when using the contralateral eye as a control and in comparing visual thalami in unilateral models of glaucoma. Translational Relevance The use of a contralateral eye as a control may confound the discovery of human-relevant mechanism and treatments in animal models. We also identify neuroinflammatory protein responses that warrant further investigation as potential disease-modifiable targets.
Collapse
Affiliation(s)
- James R. Tribble
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Eirini Kokkali
- School of Optometry and Vision Sciences, Cardiff University, Cardiff, Wales, UK
| | - Amin Otmani
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Flavia Plastino
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Emma Lardner
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Rupali Vohra
- Department of Veterinary and Animal Sciences, Pathobiological Sciences, University of Copenhagen, Denmark
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Miriam Kolko
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
- Department of Ophthalmology, Rigshospitalet-Glostrup, Copenhagen, Denmark
| | - Helder André
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - James E. Morgan
- School of Optometry and Vision Sciences, Cardiff University, Cardiff, Wales, UK
- School of Medicine, Cardiff University, Cardiff, Wales, UK
| | - Pete A. Williams
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
16
|
Transforming Growth Factor-β Signaling in Fibrotic Diseases and Cancer-Associated Fibroblasts. Biomolecules 2020; 10:biom10121666. [PMID: 33322749 PMCID: PMC7763058 DOI: 10.3390/biom10121666] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 12/08/2020] [Accepted: 12/10/2020] [Indexed: 02/06/2023] Open
Abstract
Transforming growth factor-β (TGF-β) signaling is essential in embryo development and maintaining normal homeostasis. Extensive evidence shows that TGF-β activation acts on several cell types, including epithelial cells, fibroblasts, and immune cells, to form a pro-fibrotic environment, ultimately leading to fibrotic diseases. TGF-β is stored in the matrix in a latent form; once activated, it promotes a fibroblast to myofibroblast transition and regulates extracellular matrix (ECM) formation and remodeling in fibrosis. TGF-β signaling can also promote cancer progression through its effects on the tumor microenvironment. In cancer, TGF-β contributes to the generation of cancer-associated fibroblasts (CAFs) that have different molecular and cellular properties from activated or fibrotic fibroblasts. CAFs promote tumor progression and chronic tumor fibrosis via TGF-β signaling. Fibrosis and CAF-mediated cancer progression share several common traits and are closely related. In this review, we consider how TGF-β promotes fibrosis and CAF-mediated cancer progression. We also discuss recent evidence suggesting TGF-β inhibition as a defense against fibrotic disorders or CAF-mediated cancer progression to highlight the potential implications of TGF-β-targeted therapies for fibrosis and cancer.
Collapse
|
17
|
Luis J, Eastlake K, Khaw PT, Limb GA. Galectins and their involvement in ocular disease and development. Exp Eye Res 2020; 197:108120. [PMID: 32565112 DOI: 10.1016/j.exer.2020.108120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 05/25/2020] [Accepted: 06/15/2020] [Indexed: 12/27/2022]
Abstract
Galectins are carbohydrate binding proteins with high affinity to ß-galactoside containing glycoconjugates. Understanding of the functions of galectins has grown steadily over the past decade, as a result of substantial advancements in the field of glycobiology. Galectins have been shown to be versatile molecules that participate in a range of important biological systems, including inflammation, neovascularisation and fibrosis. These processes are of particular importance in ocular tissues, where a major theme of recent research has been to divert diseases away from pathways which result in loss of function into pathways of repair and regeneration. This review summarises our current understanding of galectins in the context important ocular diseases, followed by an update on current clinical studies and future directions.
Collapse
Affiliation(s)
- Joshua Luis
- National Institute for Health Research (NIHR), Biomedical Research Centre at Moorfields Eye Hospital, NHS Foundation Trust, UCL Institute of Ophthalmology, London, EC1V 9EL, United Kingdom.
| | - Karen Eastlake
- National Institute for Health Research (NIHR), Biomedical Research Centre at Moorfields Eye Hospital, NHS Foundation Trust, UCL Institute of Ophthalmology, London, EC1V 9EL, United Kingdom
| | - Peng T Khaw
- National Institute for Health Research (NIHR), Biomedical Research Centre at Moorfields Eye Hospital, NHS Foundation Trust, UCL Institute of Ophthalmology, London, EC1V 9EL, United Kingdom
| | - G Astrid Limb
- National Institute for Health Research (NIHR), Biomedical Research Centre at Moorfields Eye Hospital, NHS Foundation Trust, UCL Institute of Ophthalmology, London, EC1V 9EL, United Kingdom
| |
Collapse
|
18
|
Tejwani S, Machiraju P, Nair AP, Ghosh A, Das RK, Ghosh A, Sethu S. Treatment of glaucoma by prostaglandin agonists and beta-blockers in combination directly reduces pro-fibrotic gene expression in trabecular meshwork. J Cell Mol Med 2020; 24:5195-5204. [PMID: 32267082 PMCID: PMC7205793 DOI: 10.1111/jcmm.15172] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 12/19/2019] [Accepted: 02/19/2020] [Indexed: 02/06/2023] Open
Abstract
Prostaglandin analogues (PG), beta-blockers (BB) or their combination (PG+BB) are used primarily to reduce the intraocular pressure (IOP) pathologically associated with glaucoma. Since, fibrosis of the trabecular meshwork (TM) is a major aetiological factor in glaucoma, we studied the effect of these drugs on fibrosis-associated gene expression in TM of primary glaucoma patients. In the present study, TM and iris of primary open-angle (n = 32) and angle-closure (n = 37) glaucoma patients were obtained surgically during trabeculectomy and categorized based on the type of IOP-lowering medications use as PG, BB or PG+BB. mRNA expression of pro-fibrotic and anti-fibrotic genes was quantified using qPCR in these tissues. The gene expression levels of pro-fibrotic genes were significantly lower in PG+BB as compared to other groups. These observations and underlying signalling validated in vitro in human TM cells also showed reduced fibrotic gene and protein expression levels following PG+BB treatment. In conclusion, it is observed that PG+BB combination rather than their lone use renders a reduced fibrotic status in TM. This further suggests that IOP-lowering medications, in combination, would also modulate fibrosis-associated molecular changes in the TM, which may be beneficial for maintaining aqueous out-flow mechanisms over the clinical treatment duration.
Collapse
Affiliation(s)
- Sushma Tejwani
- Department of Glaucoma and Cataract services, Narayana Nethralaya, Bangalore, India.,School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, India
| | - Praveen Machiraju
- GROW Research Laboratory, Narayana Nethralaya Foundation, Bangalore, India
| | - Archana Padmanabhan Nair
- GROW Research Laboratory, Narayana Nethralaya Foundation, Bangalore, India.,Manipal Academy of Higher Education, Manipal, India
| | - Anuprita Ghosh
- GROW Research Laboratory, Narayana Nethralaya Foundation, Bangalore, India
| | - Raunak Kumar Das
- Centre for Biomaterials, Cellular and Molecular Theranostics, Vellore Institute of Technology, Vellore, India
| | - Arkasubhra Ghosh
- GROW Research Laboratory, Narayana Nethralaya Foundation, Bangalore, India.,Singapore Eye Research Institute, Singapore City, Singapore
| | - Swaminathan Sethu
- GROW Research Laboratory, Narayana Nethralaya Foundation, Bangalore, India
| |
Collapse
|