1
|
Zhang T, Liu M. Cytokines in age-related eye diseases: pathogenesis and potential targets for innovative therapies. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03926-1. [PMID: 40021512 DOI: 10.1007/s00210-025-03926-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 02/12/2025] [Indexed: 03/03/2025]
Abstract
Age-related eye diseases (AREDs), such as dry eye disease (DED), age-related macular degeneration (AMD), glaucoma, and diabetic retinopathy (DR), are significant worldwide health concerns due to their rising prevalence and debilitating effects. Despite substantial research on the pathobiology of AREDs, the impact of immune-related alterations caused by aging is still not well understood. Tissue-resident cells and invading immune cells in the eye control innate responses in the event of damage or infection. However, as cells age, they gradually lose their ability to perform their protective duties and develop abnormal characteristics. Therefore, the disrupted regulation of immune responses in the eyes of older individuals enhances their vulnerability to and the intensity of eye disorders. Cytokines, immune system components, have a role in developing AREDs by contributing to inflammation. This paper examines the deficiencies in the pathogenic and therapeutic aspects of pro-inflammatory cytokines in AREDs that require further investigation in future studies.
Collapse
Affiliation(s)
- Tingting Zhang
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Xi'an Jiaotong University (Xibei Hospital), Xi'an, 710004, China
| | - Ming Liu
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Xi'an Jiaotong University (Xibei Hospital), Xi'an, 710004, China.
| |
Collapse
|
2
|
Xue Y, Ye L, Huang C, Ye H. Lights and Shadows of Cytokines in Age-Related Eye Diseases: A Narrative Literature Review. J Biochem Mol Toxicol 2025; 39:e70121. [PMID: 39756061 DOI: 10.1002/jbt.70121] [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: 10/31/2024] [Revised: 11/29/2024] [Accepted: 12/21/2024] [Indexed: 01/07/2025]
Abstract
The eye is considered to be an immune-privileged region. However, several parts of the eye have distinct mechanisms for delivering immune cells to the injury sites or even in response to aging. Although these immune responses are intended to be protective, the visual acuity can be compromised by the release of pro-inflammatory cytokines by immune cells, which induce chronic inflammation and fibrosis. Age-related eye diseases (AREDs) are the primary cause of vision impairment (VI) in the elderly, with a poor comprehension of their pathophysiology. Age-related eye diseases affect both the anterior and posterior segments, resulting in diminished quality of life and risk of irreversible blindness. Immune system dysregulation and the upregulation of pro-inflammatory cytokines have been linked to AREDs, underscoring the need to comprehend inflammation's impact on ocular disorders to enhance patient symptom management. In this framework, the PubMed database was searched using the medical subject headings (MeSH) terms "Age-related eye diseases," "dry eye syndrome," "glaucoma," "cataract," "diabetic retinopathy," "inflammation," "interleukin," and "cytokine" with the aim of overview the role of cytokines in AREDs and discuss their potential therapeutic approaches.
Collapse
Affiliation(s)
- Yuyu Xue
- Eye School of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- School of Preclinical Medicine, Chengdu University, Chengdu, Sichuan, China
| | - Lu Ye
- Department of Oncology, The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, Sichuan, China
| | - Chan Huang
- School of Preclinical Medicine, Chengdu University, Chengdu, Sichuan, China
| | - Hejiang Ye
- Department of Ophthalmology, Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| |
Collapse
|
3
|
Liang S. Role of T cell-induced autoimmune response in the pathogenesis of glaucoma. Int Ophthalmol 2024; 44:241. [PMID: 38904796 DOI: 10.1007/s10792-024-03224-4] [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: 10/06/2022] [Accepted: 06/16/2024] [Indexed: 06/22/2024]
Abstract
PURPOSE This review aims to elucidate the role of T cell-induced autoimmune responses in the pathogenesis of glaucoma, focusing on the immunological changes contributing to retinal ganglion cell (RGC) damage. METHODS A comprehensive review of recent studies examining immunological mechanisms in glaucoma was conducted. This included analyses of T cell interactions, heat shock proteins (HSPs), and resultant autoimmune responses. Key findings from experimental models and clinical observations were synthesized to present a coherent understanding of immune dynamics in glaucoma. RESULTS Glaucoma is a neurodegenerative disease marked by optic nerve atrophy and irreversible vision loss due to RGC damage. The disease is etiologically heterogeneous, with multiple risk factors and pathogenic mechanisms. Recent research highlights the dual immunomodulatory role of T cells in immune protection and injury. T cells, pre-sensitized by bacterial HSPs, can cross-react with endogenous HSPs in RGCs under stress, leading to autoimmune damage. Elevated levels of HSP autoantibodies and abnormal T cell activity have been observed in glaucoma patients, indicating a significant autoimmune component in disease progression. CONCLUSIONS T cell-induced autoimmune responses are crucial in the pathogenesis of glaucoma, contributing to RGC degeneration beyond the effects of elevated intraocular pressure. Understanding these immunological mechanisms is vital for developing targeted neuroprotective therapies for glaucoma.
Collapse
Affiliation(s)
- Shuxin Liang
- The Red Bird Program, The Hong Kong University of Science and Technology (Guangzhou), Guangzhou, Guangdong Province, China.
| |
Collapse
|
4
|
Wang D, Pu Y, Tan S, Wang X, Zeng L, Lei J, Gao X, Li H. Identification of immune-related biomarkers for glaucoma using gene expression profiling. Front Genet 2024; 15:1366453. [PMID: 38694874 PMCID: PMC11062407 DOI: 10.3389/fgene.2024.1366453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 04/03/2024] [Indexed: 05/04/2024] Open
Abstract
Introduction: Glaucoma, a principal cause of irreversible vision loss, is characterized by intricate optic neuropathy involving significant immune mechanisms. This study seeks to elucidate the molecular and immune complexities of glaucoma, aiming to improve our understanding of its pathogenesis. Methods: Gene expression profiles from glaucoma patients were analyzed to identify immune-related differentially expressed genes (DEGs). Techniques used were weighted gene co-expression network analysis (WGCNA) for network building, machine learning algorithms for biomarker identification, establishment of subclusters related to immune reactions, and single-sample gene set enrichment analysis (ssGSEA) to explore hub genes' relationships with immune cell infiltration and immune pathway activation. Validation was performed using an NMDA-induced excitotoxicity model and RT-qPCR for hub gene expression measurement. Results: The study identified 409 DEGs differentiating healthy individuals from glaucoma patients, highlighting the immune response's significance in disease progression. Immune cell infiltration analysis revealed elevated levels of activated dendritic cells, natural killer cells, monocytes, and immature dendritic cells in glaucoma samples. Three hub genes, CD40LG, TEK, and MDK, were validated as potential diagnostic biomarkers for high-risk glaucoma patients, showing increased expression in the NMDA-induced excitotoxicity model. Discussion: The findings propose the three identified immune-related genes (IRGs) as novel diagnostic markers for glaucoma, offering new insights into the disease's pathogenesis and potential therapeutic targets. The strong correlation between these IRGs and immune responses underscores the intricate role of immunity in glaucoma, suggesting a shift in the approach to its diagnosis and treatment.
Collapse
Affiliation(s)
- Dangdang Wang
- Department of Ophthalmology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for the Prevention and Treatment of Major Blinding Eye Diseases, Chongqing Eye Institute, Chongqing Branch of National Clinical Research Center for Ocular Diseases, Chongqing, China
| | - Yanyu Pu
- Department of Ophthalmology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for the Prevention and Treatment of Major Blinding Eye Diseases, Chongqing Eye Institute, Chongqing Branch of National Clinical Research Center for Ocular Diseases, Chongqing, China
| | - Sisi Tan
- Department of Ophthalmology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for the Prevention and Treatment of Major Blinding Eye Diseases, Chongqing Eye Institute, Chongqing Branch of National Clinical Research Center for Ocular Diseases, Chongqing, China
| | - Xiaochen Wang
- Department of Ophthalmology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for the Prevention and Treatment of Major Blinding Eye Diseases, Chongqing Eye Institute, Chongqing Branch of National Clinical Research Center for Ocular Diseases, Chongqing, China
| | - Lihong Zeng
- Department of Ophthalmology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for the Prevention and Treatment of Major Blinding Eye Diseases, Chongqing Eye Institute, Chongqing Branch of National Clinical Research Center for Ocular Diseases, Chongqing, China
| | - Junqin Lei
- Department of Ophthalmology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for the Prevention and Treatment of Major Blinding Eye Diseases, Chongqing Eye Institute, Chongqing Branch of National Clinical Research Center for Ocular Diseases, Chongqing, China
| | - Xi Gao
- Department of Ophthalmology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for the Prevention and Treatment of Major Blinding Eye Diseases, Chongqing Eye Institute, Chongqing Branch of National Clinical Research Center for Ocular Diseases, Chongqing, China
| | - Hong Li
- Department of Ophthalmology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for the Prevention and Treatment of Major Blinding Eye Diseases, Chongqing Eye Institute, Chongqing Branch of National Clinical Research Center for Ocular Diseases, Chongqing, China
| |
Collapse
|
5
|
Chen S, Wang N, Xiong S, Xia X. The correlation between primary open-angle glaucoma (POAG) and gut microbiota: a pilot study towards predictive, preventive, and personalized medicine. EPMA J 2023; 14:539-552. [PMID: 37605653 PMCID: PMC10439875 DOI: 10.1007/s13167-023-00336-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 07/29/2023] [Indexed: 08/23/2023]
Abstract
Background Glaucoma is the leading cause of irreversible blindness worldwide. Emerged evidence has shown that glaucoma is considered an immune system related disorder. The gut is the largest immune organ in the human body and the gut microbiota (GM) plays an irreversible role in maintaining immune homeostasis. But, how the GM influences glaucoma remains unrevealed. This study aimed at investigating the key molecules/pathways mediating the GM and the glaucoma to provide new biomarkers for future predictive, preventive, and personalized medicine. Methods Datasets from the primary open-angle glaucoma (POAG) patients (GSE138125) and datasets for target genes of GM/GM metabolites were downloaded from a public database. For GSE138125, the differentially expressed genes (DEGs) between healthy and POAG samples were identified. And the online Venn diagram tool was used to obtain the DEGs from POAG related to GM. After which GM-related DEGs were analyzed by correlation analysis, pathway enrichment analysis, and protein-protein interaction (PPI) network analysis. Human trabecular meshwork cells were used for validation, and the mRNA level of hub genes was verified by quantitative real-time polymerase chain reaction (RT-qPCR) in the in vitro glaucoma model. Results A total of 16 GM-related DEGs in POAG were identified from the above 2 datasets (9 upregulated genes and 7 downregulated genes). Pathway enrichment analysis indicated that these genes are mostly enriched in immune regulation especially macrophages-related pathways. Then 6 hub genes were identified by PPI network analysis and construction of key modules. Finally, RT-qPCR confirmed that the expression of the hub genes in the in vitro glaucoma model was consistent with the results of bioinformatics analysis of the mRNA chip. Conclusion This bioinformatic study elucidates NFKB1, IL18, KITLG, TLR9, FKBP2, and HDAC4 as hub genes for POAG and GM regulation. Immune response modulated by macrophages plays an important role in POAG and may be potential targets for future predictive, preventive, and personalized diagnosis and treatment. Supplementary Information The online version contains supplementary material available at 10.1007/s13167-023-00336-2.
Collapse
Affiliation(s)
- Si Chen
- Eye Center of Xiangya Hospital, Central South University, Changsha, 410008 Hunan China
- Hunan Key Laboratory of Ophthalmology, Changsha, Hunan China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan China
| | - Nan Wang
- Eye Center of Xiangya Hospital, Central South University, Changsha, 410008 Hunan China
- Hunan Key Laboratory of Ophthalmology, Changsha, Hunan China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan China
| | - Siqi Xiong
- Eye Center of Xiangya Hospital, Central South University, Changsha, 410008 Hunan China
- Hunan Key Laboratory of Ophthalmology, Changsha, Hunan China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan China
| | - Xiaobo Xia
- Eye Center of Xiangya Hospital, Central South University, Changsha, 410008 Hunan China
- Hunan Key Laboratory of Ophthalmology, Changsha, Hunan China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan China
| |
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
|
Wang HN, Qian WJ, Zhao GL, Li F, Miao YY, Lei B, Sun XH, Wang ZF. L- and T-type Ca 2+ channels dichotomously contribute to retinal ganglion cell injury in experimental glaucoma. Neural Regen Res 2023; 18:1570-1577. [PMID: 36571364 PMCID: PMC10075096 DOI: 10.4103/1673-5374.360277] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Retinal ganglion cell apoptotic death is the main pathological characteristic of glaucoma, which is the leading cause of irreversible blindness. Disruption of Ca2+ homeostasis plays an important role in glaucoma. Voltage-gated Ca2+ channel blockers have been shown to improve vision in patients with glaucoma. However, whether and how voltage-gated Ca2+ channels are involved in retinal ganglion cell apoptotic death are largely unknown. In this study, we found that total Ca2+ current densities in retinal ganglion cells were reduced in a rat model of chronic ocular hypertension experimental glaucoma, as determined by whole-cell patch-clamp electrophysiological recordings. Further analysis showed that L-type Ca2+ currents were downregulated while T-type Ca2+ currents were upregulated at the later stage of glaucoma. Western blot assay and immunofluorescence experiments confirmed that expression of the CaV1.2 subunit of L-type Ca2+ channels was reduced and expression of the CaV3.3 subunit of T-type Ca2+ channels was increased in retinas of the chronic ocular hypertension model. Soluble tumor necrosis factor-α, an important inflammatory factor, inhibited the L-type Ca2+ current of isolated retinal ganglion cells from control rats and enhanced the T-type Ca2+ current. These changes were blocked by the tumor necrosis factor-α inhibitor XPro1595, indicating that both types of Ca2+ currents may be mediated by soluble tumor necrosis factor-α. The intracellular mitogen-activated protein kinase/extracellular signal-regulated kinase pathway and nuclear factor kappa-B signaling pathway mediate the effects of tumor necrosis factor-α. TUNEL assays revealed that mibefradil, a T-type calcium channel blocker, reduced the number of apoptotic retinal ganglion cells in the rat model of chronic ocular hypertension. These results suggest that T-type Ca2+ channels are involved in disrupted Ca2+ homeostasis and apoptosis of retinal ganglion cells in glaucoma, and application of T-type Ca2+ channel blockers, especially a specific CaV3.3 blocker, may be a potential strategy for the treatment of glaucoma.
Collapse
Affiliation(s)
- Hong-Ning Wang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Wen-Jing Qian
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Guo-Li Zhao
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Fang Li
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yan-Ying Miao
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Bo Lei
- Institutes of Neuroscience and Third Affiliated Hospital, Henan Provincial People's Hospital, Henan Eye Institute, Henan Eye Hospital, People's Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Xing-Huai Sun
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, NHC Key Laboratory of Myopia, Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, China
| | - Zhong-Feng Wang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
8
|
Miao Y, Zhao GL, Cheng S, Wang Z, Yang XL. Activation of retinal glial cells contributes to the degeneration of ganglion cells in experimental glaucoma. Prog Retin Eye Res 2023; 93:101169. [PMID: 36736070 DOI: 10.1016/j.preteyeres.2023.101169] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 01/12/2023] [Accepted: 01/24/2023] [Indexed: 02/04/2023]
Abstract
Elevation of intraocular pressure (IOP) is a major risk factor for neurodegeneration in glaucoma. Glial cells, which play an important role in normal functioning of retinal neurons, are well involved into retinal ganglion cell (RGC) degeneration in experimental glaucoma animal models generated by elevated IOP. In response to elevated IOP, mGluR I is first activated and Kir4.1 channels are subsequently inhibited, which leads to the activation of Müller cells. Müller cell activation is followed by a complex process, including proliferation, release of inflammatory and growth factors (gliosis). Gliosis is further regulated by several factors. Activated Müller cells contribute to RGC degeneration through generating glutamate receptor-mediated excitotoxicity, releasing cytotoxic factors and inducing microglia activation. Elevated IOP activates microglia, and following morphological and functional changes, these cells, as resident immune cells in the retina, show adaptive immune responses, including an enhanced release of pro-inflammatory factors (tumor neurosis factor-α, interleukins, etc.). These ATP and Toll-like receptor-mediated responses are further regulated by heat shock proteins, CD200R, chemokine receptors, and metabotropic purinergic receptors, may aggravate RGC loss. In the optic nerve head, astrogliosis is initiated and regulated by a complex reaction process, including purines, transmitters, chemokines, growth factors and cytokines, which contributes to RGC axon injury through releasing pro-inflammatory factors and changing extracellular matrix in glaucoma. The effects of activated glial cells on RGCs are further modified by the interplay among different types of glial cells. This review is concluded by presenting an in-depth discussion of possible research directions in this field in the future.
Collapse
Affiliation(s)
- Yanying Miao
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Guo-Li Zhao
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Shuo Cheng
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Zhongfeng Wang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China.
| | - Xiong-Li Yang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
9
|
Zavarzadeh PG, Abedi Z. Novel potential drugs for the treatment of primary open-angle glaucoma using protein-protein interaction network analysis. Genomics Inform 2023; 21:e6. [PMID: 37037464 PMCID: PMC10085733 DOI: 10.5808/gi.22070] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 01/07/2023] [Indexed: 04/03/2023] Open
Abstract
Glaucoma is the second leading cause of irreversible blindness, and primary open-angle glaucoma (POAG) is the most common type. Due to inadequate diagnosis, treatment is often not administered until symptoms occur. Hence, approaches enabling earlier prediction or diagnosis of POAG are necessary. We aimed to identify novel drugs for glaucoma through bioinformatics and network analysis. Data from 36 samples, obtained from the trabecular meshwork of healthy individuals and patients with POAG, were acquired from a dataset. Next, differentially expressed genes (DEGs) were identified to construct a protein-protein interaction (PPI) network. In both stages, the genes were enriched by studying the critical biological processes and pathways related to POAG. Finally, a drug-gene network was constructed, and novel drugs for POAG treatment were proposed. Genes with p < 0.01 and |log fold change| > 0.3 (1,350 genes) were considered DEGs and utilized to construct a PPI network. Enrichment analysis yielded several key pathways that were upregulated or downregulated. For example, extracellular matrix organization, the immune system, neutrophil degranulation, and cytokine signaling were upregulated among immune pathways, while signal transduction, the immune system, extracellular matrix organization, and receptor tyrosine kinase signaling were downregulated. Finally, novel drugs including metformin hydrochloride, ixazomib citrate, and cisplatin warrant further analysis of their potential roles in POAG treatment. The candidate drugs identified in this computational analysis require in vitro and in vivo validation to confirm their effectiveness in POAG treatment. This may pave the way for understanding life-threatening disorders such as cancer.
Collapse
Affiliation(s)
- Parisima Ghaffarian Zavarzadeh
- Laboratory of Systems Biology and Bioinformatics (LBB), Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Zahra Abedi
- Laboratory of Systems Biology and Bioinformatics (LBB), Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Belvaux, Luxembourg
| |
Collapse
|
10
|
Lambuk L, Ahmad S, Sadikan MZ, Nordin NA, Kadir R, Nasir NAA, Chen X, Boer J, Plebanski M, Mohamud R. Targeting Differential Roles of Tumor Necrosis Factor Receptors as a Therapeutic Strategy for Glaucoma. Front Immunol 2022; 13:857812. [PMID: 35651608 PMCID: PMC9149562 DOI: 10.3389/fimmu.2022.857812] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 04/19/2022] [Indexed: 11/13/2022] Open
Abstract
Glaucoma is an irreversible sight-threatening disorder primarily due to elevated intraocular pressure (IOP), leading to retinal ganglion cell (RGC) death by apoptosis with subsequent loss of optic nerve fibers. A considerable amount of empirical evidence has shown the significant association between tumor necrosis factor cytokine (TNF; TNFα) and glaucoma; however, the exact role of TNF in glaucoma progression remains unclear. Total inhibition of TNF against its receptors can cause side effects, although this is not the case when using selective inhibitors. In addition, TNF exerts its antithetic roles via stimulation of two receptors, TNF receptor I (TNFR1) and TNF receptor II (TNFR2). The pro-inflammatory responses and proapoptotic signaling pathways predominantly mediated through TNFR1, while neuroprotective and anti-apoptotic signals induced by TNFR2. In this review, we attempt to discuss the involvement of TNF receptors (TNFRs) and their signaling pathway in ocular tissues with focus on RGC and glial cells in glaucoma. This review also outlines the potential application TNFRs agonist and/or antagonists as neuroprotective strategy from a therapeutic standpoint. Taken together, a better understanding of the function of TNFRs may lead to the development of a treatment for glaucoma.
Collapse
Affiliation(s)
- Lidawani Lambuk
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu, Malaysia
| | - Suhana Ahmad
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu, Malaysia
| | - Muhammad Zulfiqah Sadikan
- Centre for Neuroscience Research (NeuRon), Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh, Malaysia
| | - Nor Asyikin Nordin
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu, Malaysia
| | - Ramlah Kadir
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu, Malaysia
| | - Nurul Alimah Abdul Nasir
- Centre for Neuroscience Research (NeuRon), Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh, Malaysia
| | - Xin Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China
| | - Jennifer Boer
- School of Health and Biomedical Sciences, Royal Melbourne Institute Technology (RMIT) University, Bundoora, VIC, Australia
| | - Magdalena Plebanski
- School of Health and Biomedical Sciences, Royal Melbourne Institute Technology (RMIT) University, Bundoora, VIC, Australia
| | - Rohimah Mohamud
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu, Malaysia
| |
Collapse
|
11
|
Zhong H, Sun X. Contribution of Interleukin-17A to Retinal Degenerative Diseases. Front Immunol 2022; 13:847937. [PMID: 35392087 PMCID: PMC8980477 DOI: 10.3389/fimmu.2022.847937] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 02/25/2022] [Indexed: 12/26/2022] Open
Abstract
Retinal degenerative diseases are a leading cause of vision loss and blindness throughout the world, characterized by chronic and progressive loss of neurons and/or myelin. One of the common features of retinal degenerative diseases and central neurodegenerative diseases is chronic neuroinflammation. Interleukin-17A (IL-17A) is the cytokine most closely related to disease in its family. Accumulating evidence suggests that IL-17A plays a key role in human retinal degenerative diseases, including age-related macular degeneration, diabetic retinopathy and glaucoma. This review aims to provide an overview of the role of IL-17A participating in the pathogenesis of retinal degenerative diseases, which may open new avenues for potential therapeutic interventions.
Collapse
Affiliation(s)
- Huimin Zhong
- Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Clinical Research Center for Eye Diseases, Shanghai, China.,Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China.,Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China.,Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Xiaodong Sun
- Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Clinical Research Center for Eye Diseases, Shanghai, China.,Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China.,Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China.,Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| |
Collapse
|
12
|
Abstract
PURPOSE OF REVIEW Biomechanics is an important aspect of the complex family of diseases known as the glaucomas. Here, we review recent studies of biomechanics in glaucoma. RECENT FINDINGS Several tissues have direct and/or indirect biomechanical roles in various forms of glaucoma, including the trabecular meshwork, cornea, peripapillary sclera, optic nerve head/sheath, and iris. Multiple mechanosensory mechanisms and signaling pathways continue to be identified in both the trabecular meshwork and optic nerve head. Further, the recent literature describes a variety of approaches for investigating the role of tissue biomechanics as a risk factor for glaucoma, including pathological stiffening of the trabecular meshwork, peripapillary scleral structural changes, and remodeling of the optic nerve head. Finally, there have been advances in incorporating biomechanical information in glaucoma prognoses, including corneal biomechanical parameters and iridial mechanical properties in angle-closure glaucoma. SUMMARY Biomechanics remains an active aspect of glaucoma research, with activity in both basic science and clinical translation. However, the role of biomechanics in glaucoma remains incompletely understood. Therefore, further studies are indicated to identify novel therapeutic approaches that leverage biomechanics. Importantly, clinical translation of appropriate assays of tissue biomechanical properties in glaucoma is also needed.
Collapse
Affiliation(s)
- Babak N. Safa
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology/Emory University, Atlanta GA, USA
| | - Cydney A. Wong
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology/Emory University, Atlanta GA, USA
| | - Jungmin Ha
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology/Emory University, Atlanta GA, USA
| | - C. Ross Ethier
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology/Emory University, Atlanta GA, USA
| |
Collapse
|
13
|
Leung DYL, Tham CC. Normal-tension glaucoma: Current concepts and approaches-A review. Clin Exp Ophthalmol 2022; 50:247-259. [PMID: 35040248 DOI: 10.1111/ceo.14043] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 01/09/2022] [Accepted: 01/11/2022] [Indexed: 12/19/2022]
Abstract
Normal tension glaucoma (NTG) has remained a challenging disease. We review, from an epidemiological perspective, why we should redefine normality, act earlier at lower pre-treatment intraocular pressure (IOP) level, and the role of ocular perfusion pressures, noting that perfusion is affected by defective vascular bed autoregulation and endothelial dysfunction. The correlation of silent cerebral infarcts (SCI) and NTG may indicate that NTG belongs to a wider spectrum of small vessel diseases (SVD), with its main pathology being also on vascular endothelium. Epidemiological studies also suggested that vascular geometry, such as fractal dimension, may affect perfusion efficiency, occurrence of SCI, SVD and glaucoma. Artificial intelligence with deep learning, may help predicting NTG progression from vascular geometry. Finally, we review latest evidence on the role of minimally-invasive glaucoma surgery, lasers, and newer drugs. We conclude that IOP is not the only modifiable risk factors as, many vascular risk factors are readily modifiable.
Collapse
Affiliation(s)
- Dexter Y L Leung
- Department of Ophthalmology & Visual Sciences, The Chinese University of Hong Kong, Kowloon, Hong Kong SAR, China
- Department of Ophthalmology, Hong Kong Sanatorium & Hospital, Happy Valley, Hong Kong SAR, China
| | - Clement C Tham
- Department of Ophthalmology & Visual Sciences, The Chinese University of Hong Kong, Kowloon, Hong Kong SAR, China
- Hong Kong Eye Hospital, Kowloon, Hong Kong SAR, China
- Lam Kin Chung . Jet King-Shing Ho Glaucoma Treatment and Research Centre, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
14
|
Alapati T, Sagal KM, Gudiseva HV, Pistilli M, Pyfer M, Chavali VRM, O’Brien JM. Evaluating TNF-α and Interleukin-2 (IL-2) Levels in African American Primary Open-Angle Glaucoma Patients. Genes (Basel) 2021; 13:genes13010054. [PMID: 35052396 PMCID: PMC8774833 DOI: 10.3390/genes13010054] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 12/19/2021] [Accepted: 12/23/2021] [Indexed: 12/30/2022] Open
Abstract
Purpose: To establish if SNPs in TNF-α and IL-2 genes are associated with Primary Open-Angle Glaucoma (POAG) in African Americans (AA). We also determined whether plasma TNF-α and IL-2 levels could serve as biomarkers for POAG in African Americans using sandwich enzyme-linked immunosorbent assay. Methods: A single SNP association analysis was performed to investigate the association between potential gene variants in TNF-α and IL-2 genes and POAG in the AA population. Plasma samples from 190 African Americans (72 from normal subjects and 118 POAG cases) were obtained for TNF- α studies and 367 samples (135 from normal subjects and 232 from POAG cases) were obtained for IL-2 studies. TNF-α levels and IL-2 levels were measured by sandwich enzyme-linked immunosorbent assays (ELISA) and analyzed to see if they reached significance in cases with POAG and endophenotypes when compared to normal subjects. Results: The SNP, rs1800630, in TNF-α gene was found to be marginally associated with POAG. SNPs in IL-2 gene were not associated with POAG in the case-control analysis. No significant difference was found between TNF-α levels and IL-2 levels in normal and POAG case subjects in our study. IL-2 levels were inversely correlated with high IOP in POAG cases. Conclusions: Although we found a marginal SNP association of TNF-α, assessing the expression levels of TNF-α and IL-2 may serve as promising biomarkers for African American POAG. Further investigation is needed to determine if POAG can be subdivided into more specified cohorts of the disease, which may affect plasma cytokine levels differently.
Collapse
|
15
|
Proteomic Analysis of Retinal Tissue in an S100B Autoimmune Glaucoma Model. BIOLOGY 2021; 11:biology11010016. [PMID: 35053014 PMCID: PMC8773367 DOI: 10.3390/biology11010016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 12/15/2021] [Accepted: 12/20/2021] [Indexed: 12/13/2022]
Abstract
Glaucoma is a neurodegenerative disease that leads to damage of retinal ganglion cells and the optic nerve. Patients display altered antibody profiles and increased antibody titer, e.g., against S100B. To identify the meaning of these antibodies, animals were immunized with S100B. Retinal ganglion cell loss, optic nerve degeneration, and increased glial cell activity were noted. Here, we aimed to gain more insights into the pathophysiology from a proteomic point of view. Hence, rats were immunized with S100B, while controls received sodium chloride. After 7 and 14 days, retinae were analyzed through mass spectrometry and immunohistology. Using data-independent acquisition-based mass spectrometry, we identified more than 1700 proteins on a high confidence level for both study groups, respectively. Of these 1700, 43 proteins were significantly altered in retinae after 7 days and 67 proteins revealed significant alterations at 14 days. For example, α2-macroglobulin was found significantly increased not only by mass spectrometry analysis, but also with immunohistological staining in S100B retinae at 7 and 14 days. All in all, the identified proteins are often associated with the immune system, such as heat shock protein 60. Once more, these data underline the important role of immunological factors in glaucoma pathogenesis.
Collapse
|
16
|
Wu Y, Szymanska M, Hu Y, Fazal MI, Jiang N, Yetisen AK, Cordeiro MF. Measures of disease activity in glaucoma. Biosens Bioelectron 2021; 196:113700. [PMID: 34653715 DOI: 10.1016/j.bios.2021.113700] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 10/01/2021] [Accepted: 10/08/2021] [Indexed: 12/13/2022]
Abstract
Glaucoma is the leading cause of irreversible blindness globally which significantly affects the quality of life and has a substantial economic impact. Effective detective methods are necessary to identify glaucoma as early as possible. Regular eye examinations are important for detecting the disease early and preventing deterioration of vision and quality of life. Current methods of measuring disease activity are powerful in describing the functional and structural changes in glaucomatous eyes. However, there is still a need for a novel tool to detect glaucoma earlier and more accurately. Tear fluid biomarker analysis and new imaging technology provide novel surrogate endpoints of glaucoma. Artificial intelligence is a post-diagnostic tool that can analyse ophthalmic test results. A detail review of currently used clinical tests in glaucoma include intraocular pressure test, visual field test and optical coherence tomography are presented. The advanced technologies for glaucoma measurement which can identify specific disease characteristics, as well as the mechanism, performance and future perspectives of these devices are highlighted. Applications of AI in diagnosis and prediction in glaucoma are mentioned. With the development in imaging tools, sensor technologies and artificial intelligence, diagnostic evaluation of glaucoma must assess more variables to facilitate earlier diagnosis and management in the future.
Collapse
Affiliation(s)
- Yue Wu
- Department of Surgery and Cancer, Imperial College London, South Kensington, London, United Kingdom; Department of Chemical Engineering, Imperial College London, South Kensington, London, United Kingdom
| | - Maja Szymanska
- The Imperial College Ophthalmic Research Group (ICORG), Imperial College London, London, United Kingdom
| | - Yubing Hu
- Department of Chemical Engineering, Imperial College London, South Kensington, London, United Kingdom.
| | - M Ihsan Fazal
- The Imperial College Ophthalmic Research Group (ICORG), Imperial College London, London, United Kingdom
| | - Nan Jiang
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Ali K Yetisen
- Department of Chemical Engineering, Imperial College London, South Kensington, London, United Kingdom
| | - M Francesca Cordeiro
- The Imperial College Ophthalmic Research Group (ICORG), Imperial College London, London, United Kingdom; The Western Eye Hospital, Imperial College Healthcare NHS Trust (ICHNT), London, United Kingdom; Glaucoma and Retinal Neurodegeneration Group, Department of Visual Neuroscience, UCL Institute of Ophthalmology, London, United Kingdom.
| |
Collapse
|
17
|
Arrigo A, Aragona E, Saladino A, Arrigo D, Fantaguzzi F, Battaglia Parodi M, Bandello F. Cognitive Dysfunctions in Glaucoma: An Overview of Morpho-Functional Mechanisms and the Impact on Higher-Order Visual Function. Front Aging Neurosci 2021; 13:747050. [PMID: 34690746 PMCID: PMC8526892 DOI: 10.3389/fnagi.2021.747050] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 08/27/2021] [Indexed: 12/11/2022] Open
Abstract
Background: Glaucoma is a chronic, vision-threatening disease, and a major cause of legal blindness. The current view is no longer limited to the progressive optic nerve injury, since growing evidence strongly support the interpretation of glaucoma as a complex neurodegenerative disease. However, the precise pathogenic mechanisms leading to the onset and progression of central nervous system (CNS) impairment, and the functional consequences of this damage, are still partially understood. The main aim of this review is to provide a complete and updated overview of the current knowledge regarding the CNS involvement in glaucoma, and the possible therapeutic perspectives. Methods: We made a careful survey of the current literature reporting all the relevant findings related to the cognitive dysfunctions occurring in glaucoma, with specific remarks dedicated on the higher-order visual function impairment and the possible employment of neuroprotective agents. Results: The current literature strongly support the interpretation of glaucoma as a multifaceted chronic neurodegenerative disease, widely affecting the CNS. The cognitive impairment may vary in terms of higher-order functions involvement and in the severity of the degeneration. Although several neuroprotective agents are currently available, the development of new molecules represents a major topic of investigation for future clinical trials. Conclusions: Glaucoma earned the right to be fully considered a neurodegenerative disease. Glaucomatous patients may experience a heterogeneous set of visual and cognitive symptoms, progressively deteriorating the quality of life. Neuroprotection is nowadays a necessary therapeutic goal and a future promising way to preserve visual and cognitive functions, thus improving patients' quality of life.
Collapse
Affiliation(s)
- Alessandro Arrigo
- Department of Ophthalmology, Scientific Institute San Raffaele Hospital, Milan, Italy
| | - Emanuela Aragona
- Department of Ophthalmology, Scientific Institute San Raffaele Hospital, Milan, Italy
| | - Andrea Saladino
- Department of Ophthalmology, Scientific Institute San Raffaele Hospital, Milan, Italy
| | - Davide Arrigo
- School of Medicine, University of Messina, Messina, Italy
| | - Federico Fantaguzzi
- Department of Ophthalmology, Scientific Institute San Raffaele Hospital, Milan, Italy
| | | | - Francesco Bandello
- Department of Ophthalmology, Scientific Institute San Raffaele Hospital, Milan, Italy
| |
Collapse
|
18
|
Ko KW, Milbrandt J, DiAntonio A. SARM1 acts downstream of neuroinflammatory and necroptotic signaling to induce axon degeneration. J Cell Biol 2021; 219:151915. [PMID: 32609299 PMCID: PMC7401797 DOI: 10.1083/jcb.201912047] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 04/08/2020] [Accepted: 04/27/2020] [Indexed: 12/29/2022] Open
Abstract
Neuroinflammation and necroptosis are major contributors to neurodegenerative disease, and axon dysfunction and degeneration is often an initiating event. SARM1 is the central executioner of pathological axon degeneration. Here, we demonstrate functional and mechanistic links among these three pro-degenerative processes. In a neuroinflammatory model of glaucoma, TNF-α induces SARM1-dependent axon degeneration, oligodendrocyte loss, and subsequent retinal ganglion cell death. TNF-α also triggers SARM1-dependent axon degeneration in sensory neurons via a noncanonical necroptotic signaling mechanism. MLKL is the final executioner of canonical necroptosis; however, in axonal necroptosis, MLKL does not directly trigger degeneration. Instead, MLKL induces loss of the axon survival factors NMNAT2 and STMN2 to activate SARM1 NADase activity, which leads to calcium influx and axon degeneration. Hence, these findings define a specialized form of axonal necroptosis. The demonstration that neuroinflammatory signals and necroptosis can act locally in the axon to stimulate SARM1-dependent axon degeneration identifies a therapeutically targetable mechanism by which neuroinflammation can stimulate axon loss in neurodegenerative disease.
Collapse
Affiliation(s)
- Kwang Woo Ko
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO
| | - Jeffrey Milbrandt
- Department of Genetics, Washington University School of Medicine, St. Louis, MO.,Needleman Center for Neurometabolism and Axonal Therapeutics, Washington University School of Medicine, St. Louis, MO
| | - Aaron DiAntonio
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO.,Needleman Center for Neurometabolism and Axonal Therapeutics, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
19
|
Chen J, Sun J, Yu H, Huang P, Zhong Y. Evaluation of the Effectiveness of a Chronic Ocular Hypertension Mouse Model Induced by Intracameral Injection of Cross-Linking Hydrogel. Front Med (Lausanne) 2021; 8:643402. [PMID: 33829024 PMCID: PMC8019751 DOI: 10.3389/fmed.2021.643402] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 02/10/2021] [Indexed: 12/13/2022] Open
Abstract
Background: Glaucoma is an irreversible and blinding neurodegenerative disease that is characterized by progressive loss of retinal ganglion cells. The current animal models of glaucoma fail to provide a chronic elevated intraocular pressure and cannot maintain the optical media clarity for a long time, which brings some difficulties to the study of glaucoma. Here, we developed a new chronic ocular hypertension model of mice induced by cross-linking hydrogel intracameral injection. Methods: C57BL/6J mice aged 6–8 weeks were randomly divided into the control group and the operation group. The mice of the operation group were injected with cross-linking hydrogel to induce ocular hypertension. Intraocular pressure was measured preoperatively, 3 days after surgery, and weekly until the end of the study. Flash visual evoked potential (F-VEP) was used to observe optic nerve function at different times (preoperatively and 2, 4, and 6 weeks) after chronic ocular hypertension (COH). Retinal TNF-α, IL-1β, and IL-17A protein expression were measured by western blotting in the control group and in mice at 2, 4, and 6 weeks after COH. Microglial cell activation was evaluated by immunofluorescence staining and western blotting. Apoptosis and loss of retinal ganglion cells after 2, 4, and 6 weeks of intracameral injection of cross-linking hydrogel were observed by the TUNEL assay and Brn3a protein labeling. The loss of optic nerve axons in COH mice was evaluated by neurofilament heavy polypeptide protein labeling. Results: Intracameral injection of the cross-linking hydrogel induces increased intraocular pressure (IOP) to a mean value of 19.3 ± 4.1 mmHg, which was sustained for at least 8 weeks. A significant difference in IOP was noted between COH mice and sham-operation mice (p < 0.0001). The success rate was 75%. The average amplitude of F-VEP in mice with COH was reduced (p = 0.0149, 0.0012, and 0.0009 at 2, 4, and 6 weeks after COH vs. the control group, respectively), and the average latent period in mice with COH was longer (p = 0.0290, <0.0001, and <0.0001 at 2, 4, and 6 weeks after COH vs. the control group, respectively) compared with that in the control group. TNF-α, IL-1β, IL-17A, Iba-1, and CD68 protein expression increased in COH mice. During the processing of COH, the number of microglial cells increased along with cellular morphological changes of rounder bodies and thicker processes compared with the control group. Apoptosis of retinal ganglion cells (RGCs) was clearly observed in mice at 2, 4, and 6 weeks after COH (p = 0.0061, 0.0012, <0.0001, and 0.0371 at 2, 4, and 6 weeks after COH vs. the control group, respectively). The RGC density decreased significantly in the COH mice compared with the control group (p = 0.0042, 0.0036, and <0.0001 at 2, 4, and 6 weeks after COH vs. the control group, respectively). There was a significant loss of optic nerve axons in mice after intracameral injection of cross-linking hydrogel (p = 0.0095, 0.0002, and <0.0001 at 2, 4, and 6 weeks after COH vs. the control group, respectively). Conclusions: A single intracameral injection of cross-linking hydrogel can effectively induce chronic ocular hypertension in mice, which causes progressive loss of retinal ganglion cells, increased expression levels of inflammatory cytokines and microglial cell activation, and deterioration of optic nerve function.
Collapse
Affiliation(s)
- Junjue Chen
- Department of Ophthalmology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Sun
- Department of Ophthalmology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huan Yu
- Department of Ophthalmology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ping Huang
- Shanghai Key Laboratory for Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital Affiliated Medical School, Shanghai Jiaotong University, Shanghai, China
| | - Yisheng Zhong
- Department of Ophthalmology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
20
|
Effect of depressive symptom and depressive disorder on glaucoma incidence in elderly. Sci Rep 2021; 11:5888. [PMID: 33723349 PMCID: PMC7961135 DOI: 10.1038/s41598-021-85380-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 02/18/2021] [Indexed: 12/27/2022] Open
Abstract
Although depression and glaucoma share several common pathophysiology, the risk of glaucoma in patients with depression has not been reported. Thus, we investigated the effect of depressive symptom and depressive disorder on glaucoma incidence. In this nationwide population-based cohort study, all subjects receiving the National Screening Program at the age of 66 during 2009-2014 were included. These subjects were divided into depression group and no depression group based on subjective depressive symptoms and clinically diagnosed depressive disorder and were tracked until 2017 for development of glaucoma. Of the 922,769 subjects included in the study, 191,636 (20.77%) subjects were categorized as depression group. Subjects with depression showed increased hazard of developing glaucoma (adjusted HR = 1.12[95% confidence interval (CI), 1.09-1.15]) than those without depression. The risk of glaucoma increased sequentially from those with no depression to those with subjective depressive symptom (adjusted HR = 1.09[95% CI, 1.06-1.13]), those with clinically diagnosed depressive disorder (adjusted HR = 1.23[95% CI, 1.14-1.32]), and those with both subjective depressive symptom and clinically diagnosed depressive disorder (adjusted HR = 1.36[95% CI, 1.22-1.52]). Our analyses suggest that individuals with depression had a greater risk of developing glaucoma than those without depression. Subjective depressive symptoms and clinically diagnosed depressive disorder independently and synergistically increased the risk of glaucoma incidence.
Collapse
|
21
|
Samelska K, Zaleska-Żmijewska A, Bałan B, Grąbczewski A, Szaflik JP, Kubiak AJ, Skopiński P. Immunological and molecular basics of the primary open angle glaucoma pathomechanism. Cent Eur J Immunol 2021; 46:111-117. [PMID: 33897292 PMCID: PMC8056342 DOI: 10.5114/ceji.2021.104328] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 10/26/2020] [Indexed: 02/07/2023] Open
Abstract
Glaucoma is a degenerative process of the optic nerve. Increased intraocular pressure is believed to be the main factor leading to the glaucomatous damage. The in vitro and in vivo animal glaucoma research models provide insight into the molecular changes in the retina in response to the injury factor. The damage is a complex process incorporating molecular and immunological changes. Such changes involve NF kB activity and complement activation. The processes affect the human antigen, JNK, MAPK, p53, MT2 and DBA/2J molecular pathways, activate the autophagy processes and compromise neuroprotective mechanisms. Activation and inhibition of immunological responses contribute to cell injury. The immunological mechanisms of glaucomatous degeneration include glial response, the complement, tumor necrosis factor α (TNF-α) pathways and toll-like receptors athways. Oxidative stress and excitotoxicity are factors contributing to cell death in glaucoma. The authors present an up-to-date review of the mechanisms involved and update on research focusing on a possible innovative glaucoma treatment.
Collapse
Affiliation(s)
- Katarzyna Samelska
- SPKSO Ophthalmic University Hospital, Warsaw, Poland
- Department of Ophthalmology, Medical University of Warsaw, Warsaw, Poland
| | - Anna Zaleska-Żmijewska
- SPKSO Ophthalmic University Hospital, Warsaw, Poland
- Department of Ophthalmology, Medical University of Warsaw, Warsaw, Poland
| | - Barbara Bałan
- Department of Immunology Biochemistry and Nutrition, Medical University of Warsaw, Warsaw, Poland
| | | | - Jacek Paweł Szaflik
- SPKSO Ophthalmic University Hospital, Warsaw, Poland
- Department of Ophthalmology, Medical University of Warsaw, Warsaw, Poland
| | | | - Piotr Skopiński
- SPKSO Ophthalmic University Hospital, Warsaw, Poland
- Department of Histology and Embryology, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
22
|
Abstract
The prognosis of going blind is very stressful for patients diagnosed with "glaucoma". Worries and fear of losing independence is a constant mental burden, with secondary risks of depression and social isolation. But stress is not only a result of glaucoma but also a possible cause (risk factor). This should not be surprising, given that chronic stress can trigger "psychosomatic" organ dysfunctions anywhere in the body. Why should the organ "eye" be an exception? Indeed, glaucoma patients often suspect that severe emotional stress caused their visual field loss or "foggy vision". The hypothesis that stress is a possible cause of glaucoma is supported by different observations: (i) acute and chronic stress increases intraocular pressure and (ii) long-term stress can lead to vascular dysregulation of the microcirculation in the eye and brain ("Flammer's syndrome"), leading to partial hypoxia and hypoglycaemia (hypo-metabolism). Even if nerve cells do not die, they may then become inactive ("silent" neurons). (iii) Degenerative changes have been reported in the brain of glaucoma patients, affecting not only anterograde or transsynaptic areas of the central visual pathway, but degeneration is also found (iv) in brain areas involved in emotional appraisal and the physiological regulation of stress hormones. There are also psychological hints indicating that stress is a cause of glaucoma: (v) Glaucoma patients with Flammer's syndrome show typical personality traits that are associated with low stress resilience: they often have cold hands or feet, are ambitious (professionally successful), perfectionistic, obsessive, brooding and worrying a lot. (vi) If stress hormone levels and inflammation parameters are reduced in glaucoma patients by relaxation with meditation, this correlates with normalisation of intraocular pressure, and yet another clue is that (vii) visual field improvements after non-invasive current stimulation therapy, that are known to improve circulation and neuronal synchronisation, are much most effective in patients with stress resilient personalities. An appreciation of stress as a "cause" of glaucoma suggests that in addition to standard therapy (i) stress reduction through relaxation techniques should be recommended (e.g. meditation), and (ii) self-medication compliance should not be induced by kindling anxiety and worries with negative communication ("You will go blind!"), but communication should be positive ("The prognosis is optimistic").
Collapse
Affiliation(s)
- Bernhard A Sabel
- Otto-von-Guericke Universität Magdeburg, Institut für Medizinische Psychologie, Deutschland
| | - Luisa Lehnigk
- Otto-von-Guericke Universität Magdeburg, Institut für Medizinische Psychologie, Deutschland
| |
Collapse
|
23
|
Harder JM, Williams PA, Braine CE, Yang HS, Thomas JM, Foxworth NE, John SWM, Howell GR. Complement peptide C3a receptor 1 promotes optic nerve degeneration in DBA/2J mice. J Neuroinflammation 2020; 17:336. [PMID: 33176797 PMCID: PMC7656757 DOI: 10.1186/s12974-020-02011-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 10/28/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The risk of glaucoma increases significantly with age and exposure to elevated intraocular pressure, two factors linked with neuroinflammation. The complement cascade is a complex immune process with many bioactive end-products, including mediators of inflammation. Complement cascade activation has been shown in glaucoma patients and models of glaucoma. However, the function of complement-mediated inflammation in glaucoma is largely untested. Here, the complement peptide C3a receptor 1 was genetically disrupted in DBA/2J mice, an ocular hypertensive model of glaucoma, to test its contribution to neurodegeneration. METHODS A null allele of C3ar1 was backcrossed into DBA/2J mice. Development of iris disease, ocular hypertension, optic nerve degeneration, retinal ganglion cell activity, loss of RGCs, and myeloid cell infiltration in C3ar1-deficient and sufficient DBA/2J mice were compared across multiple ages. RNA sequencing was performed on microglia from primary culture to determine global effects of C3ar1 on microglia gene expression. RESULTS Deficiency in C3ar1 lowered the risk of degeneration in ocular hypertensive mice without affecting intraocular pressure elevation at 10.5 months of age. Differences were found in the percentage of mice affected, but not in individual characteristics of disease progression. The protective effect of C3ar1 deficiency was then overcome by additional aging and ocular hypertensive injury. Microglia and other myeloid-derived cells were the primary cells identified that express C3ar1. In the absence of C3ar1, microglial expression of genes associated with neuroinflammation and other immune functions were differentially expressed compared to WT. A network analysis of these data suggested that the IL10 signaling pathway is a major interaction partner of C3AR1 signaling in microglia. CONCLUSIONS C3AR1 was identified as a damaging neuroinflammatory factor. These data help suggest complement activation causes glaucomatous neurodegeneration through multiple mechanisms, including inflammation. Microglia and infiltrating myeloid cells expressed high levels of C3ar1 and are the primary candidates to mediate its effects. C3AR1 appeared to be a major regulator of microglia reactivity and neuroinflammatory function due to its interaction with IL10 signaling and other immune related pathways. Targeting myeloid-derived cells and C3AR1 signaling with therapies is expected to add to or improve neuroprotective therapeutic strategies.
Collapse
Affiliation(s)
| | - Pete A Williams
- The Jackson Laboratory, Bar Harbor, ME, USA
- Division of Eye and Vision, Department of Clinical Neuroscience, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Catherine E Braine
- The Jackson Laboratory, Bar Harbor, ME, USA
- Zuckerman Mind Brain Behavior Institute, New York, NY, USA
| | | | | | | | - Simon W M John
- The Jackson Laboratory, Bar Harbor, ME, USA.
- Department of Ophthalmology, Tufts University of Medicine, Boston, MA, USA.
- Howard Hughes Medical Institute, Department of Ophthalmology, Columbia University Medical Center, and Zuckerman Mind Brain Behavior Institute, New York, NY, USA.
| | - Gareth R Howell
- The Jackson Laboratory, Bar Harbor, ME, USA.
- Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA.
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME, USA.
| |
Collapse
|
24
|
Abstract
Glaucoma is a chronic progressive optic neuropathy that causes irreversible loss of visual functions. From the clinical point of view, normal-tension glaucoma (NTG) is regarded in Russian taxonomy as a clinical form of standard primary open-angle glaucoma in which the IOP values stay within the normal range, but the typical progressive visual functions loss is still present. The results of the latest studies put in question the traditional views of NTG pathophysiology that are based solely on intraocular pressure values. New capabilities of diagnostic visualization of central nervous system have considerably broadened our knowledge of the NTG development mechanisms. This article reviews current understanding of the pathogenesis of NTG and its connection to vascular and immune factors, translaminar pressure difference etc. The review also considers the relationship between glaucoma and cognitive defects associated with Alzheimer's and Parkinson's diseases.
Collapse
Affiliation(s)
- S Yu Petrov
- Research Institute of Eye Diseases, Moscow, Russia
| |
Collapse
|
25
|
Jabbehdari S, Chen JL, Vajaranant TS. Effect of dietary modification and antioxidant supplementation on intraocular pressure and open-angle glaucoma. Eur J Ophthalmol 2020; 31:1588-1605. [PMID: 33008269 DOI: 10.1177/1120672120960337] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Primary open-angle glaucoma (POAG) is an age-dependent, intraocular pressure (IOP)-related degeneration of the retinal ganglion cells (RGC). At present, IOP is the only modifiable factor that has been identified to prevent glaucomatous vision loss. Though the pathogenesis of glaucomatous optic neuropathy is still not well understood, increasing evidence suggests oxidative stress may contribute to the induction and progression of glaucoma. Furthermore, antioxidant use may be protective against glaucoma through various mechanisms, including reducing IOP, preserving vascular health, and preventing ganglion cell loss. This article provides a comprehensive review of the effect of oxidative stress, diet, and antioxidant therapy on IOP and open-angle glaucoma.
Collapse
Affiliation(s)
- Sayena Jabbehdari
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Judy L Chen
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | | |
Collapse
|
26
|
Gassel CJ, Reinehr S, Gomes SC, Dick HB, Joachim SC. Preservation of optic nerve structure by complement inhibition in experimental glaucoma. Cell Tissue Res 2020; 382:293-306. [PMID: 32676862 PMCID: PMC8285355 DOI: 10.1007/s00441-020-03240-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 06/04/2020] [Indexed: 01/09/2023]
Abstract
Glaucoma is characterized by a progressive damage of the retina and the optic nerve. Despite a huge research interest, the exact pathomechanisms are still unknown. In the experimental autoimmune glaucoma model, rats develop glaucoma-like damage of the retina and the optic nerve after immunization with an optic nerve antigen homogenate (ONA). An early activation of the complement system, even before optic nerve degeneration, was reported in this model. Here, we investigated the effects of a monoclonal antibody against complement factor C5 on optic nerves. Rats were immunized with ONA and compared to controls. In one eye of some ONA animals, the antibody against C5 was intravitreally injected (15 μmol: ONA + C5-I or 25 μmol: ONA + C5-II) before immunization and then every 2 weeks. After 6 weeks, optic nerves were processed for histology (n = 6/group). These analyses demonstrated that the intravitreal therapy reduced the depositions of the membrane attack complex compared to ONA animals (ONA + C5-I: p = 0.005; ONA + C5-II: p = 0.002). Cellular infiltration was significantly reduced in the ONA + C5-I group (p = 0.003), but not in ONA + C5-II tissues (p = 0.41). Furthermore, SMI-32 staining revealed that neurofilament was preserved in both treatment groups compared to ONA optic nerves (both p = 0.002). A decreased amount of microglia was found in treated animals in comparison to the ONA group (ONA + C5-I: p = 0.03; ONA + C5-II: p = 0.009). We observed, for the first time, that a complement system inhibition could prevent optic nerve damage in an autoimmune glaucoma model. Therefore, complement inhibition could serve as a new therapeutic tool for glaucoma.
Collapse
Affiliation(s)
- Caroline J Gassel
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, In der Schornau 23-25, 44892, Bochum, Germany
| | - Sabrina Reinehr
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, In der Schornau 23-25, 44892, Bochum, Germany
| | - Sara C Gomes
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, In der Schornau 23-25, 44892, Bochum, Germany
| | - H Burkhard Dick
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, In der Schornau 23-25, 44892, Bochum, Germany
| | - Stephanie C Joachim
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, In der Schornau 23-25, 44892, Bochum, Germany.
| |
Collapse
|
27
|
Abstract
Glaucoma is characterized by retinal ganglion cell (RGC) neurodegeneration. Elevated intraocular pressure (IOP) is a major risk factor however, mechanisms independent of IOP play a role in RGC pathology. Both antibodies and CD4 T-cells as well as microbiota take part in the pathogenesis of both glaucoma and rheumatoid arteritis (RA).Heat shock proteins (HSPs) which originate in bacteria cross-react with RCG epitopes and were involved in rat model of retinal injury. Enhanced expression of HSPs in the retina was associated with glaucoma-like neuropathology and previous studies have also suggested a pathogenic role for HSPs in RA. In view of these data we suggest that glaucoma should be included in the spectrum of autoimmune diseases and that proven medications for RA should be adopted as an innovative IOP -independent therapeutic strategy for glaucoma.
Collapse
|
28
|
Reinehr S, Gomes SC, Gassel CJ, Asaad MA, Stute G, Schargus M, Dick HB, Joachim SC. Intravitreal Therapy Against the Complement Factor C5 Prevents Retinal Degeneration in an Experimental Autoimmune Glaucoma Model. Front Pharmacol 2019; 10:1381. [PMID: 31849650 PMCID: PMC6901014 DOI: 10.3389/fphar.2019.01381] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 10/30/2019] [Indexed: 12/18/2022] Open
Abstract
In glaucoma, studies revealed an involvement of the complement system. In an experimental autoimmune glaucoma model, immunization with an optic nerve homogenate antigen (ONA) led to retinal ganglion cell (RGC) loss, while intraocular pressure (IOP) remained unchanged. Here, we investigated the therapeutic effect of a complement system inhibition in this model. Hence, rats were immunized with ONA and compared to controls. In one eye of the ONA animals, an antibody against complement factor C5 was intravitreally injected (15 μmol: ONA+C5-I or 25 μmol: ONA+C5-II) before immunization and then every two weeks. IOP was measured weekly. After 6 weeks, spectral-domain optical coherence tomographies (SD-OCT), electroretinograms (ERG), immunohistochemistry, and quantitative real-time PCR analyses were performed. IOP and retinal thickness remained unchanged within all groups. The a-wave amplitudes were not altered in the ONA and ONA+C5-I groups, whereas a decrease was noted in ONA+C5-II animals (p < 0.05). ONA immunization provoked a significant decrease of the b-wave amplitude (p < 0.05), which could be preserved in ONA+C5-I, but not in ONA+C5-II animals. ONA animals showed a loss of RGCs (p = 0.001), while ONA+C5-I and ONA+C5-II retinae had similar cell counts as controls. A significant downregulation of apoptotic Bax/Bcl2 mRNA was noted in ONA+C5-I retinae (p = 0.02). Significantly more C3+ and MAC+ cells were observed in ONA animals (p < 0.001). The amount of C3+ cells in both treatment groups was significantly increased (p < 0.01), while the number of MAC+ cells in the treated retinas did not differ from controls. The number of activated microglia cells remained unchanged in ONA animals, but was increased in the treatment groups (p < 0.05). Recoverin+ cells were diminished in ONA animals (p = 0.049), but not in treated ones. Rho mRNA was downregulated in ONA and in ONA+C5-II retinas (both p = 0.014). Less opsin+ cones were observed in ONA animals (p = 0.009), but not in the treated groups. Our results indicate that the C5 antibody inhibits activation of the complement system, preventing the loss of retinal function as well as RGC, cone bipolar, and photoreceptor loss. Therefore, this approach might be a suitable new treatment for glaucoma patients, in which immune dysregulation plays an important factor for the development and progression of glaucoma.
Collapse
Affiliation(s)
- Sabrina Reinehr
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Sara C Gomes
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Caroline J Gassel
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | - M Ali Asaad
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Gesa Stute
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Marc Schargus
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany.,Department of Ophthalmology, University Hospital Duesseldorf, Heinrich-Heine-University Duesseldorf, Duesseldorf, Germany
| | - H Burkhard Dick
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Stephanie C Joachim
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| |
Collapse
|
29
|
Tsai T, Reinehr S, Maliha AM, Joachim SC. Immune Mediated Degeneration and Possible Protection in Glaucoma. Front Neurosci 2019; 13:931. [PMID: 31543759 PMCID: PMC6733056 DOI: 10.3389/fnins.2019.00931] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 08/19/2019] [Indexed: 12/13/2022] Open
Abstract
The underlying pathomechanisms for glaucoma, one of the most common causes of blindness worldwide, are still not identified. In addition to increased intraocular pressure (IOP), oxidative stress, excitotoxicity, and immunological processes seem to play a role. Several pharmacological or molecular/genetic methods are currently investigated as treatment options for this disease. Altered autoantibody levels were detected in serum, aqueous humor, and tissue sections of glaucoma patients. To further analyze the role of the immune system, an IOP-independent, experimental autoimmune glaucoma (EAG) animal model was developed. In this model, immunization with ocular antigens leads to antibody depositions, misdirected T-cells, retinal ganglion cell death and degeneration of the optic nerve, similar to glaucomatous degeneration in patients. Moreover, an activation of the complement system and microglia alterations were identified in the EAG as well as in ocular hypertension models. The inhibition of these factors can alleviate degeneration in glaucoma models with and without high IOP. Currently, several neuroprotective approaches are tested in distinct models. It is necessary to have systems that cover underlying pathomechanisms, but also allow for the screening of new drugs. In vitro models are commonly used, including single cell lines, mixed-cultures, and even organoids. In ex vivo organ cultures, pathomechanisms as well as therapeutics can be investigated in the whole retina. Furthermore, animal models reveal insights in the in vivo situation. With all these models, several possible new drugs and therapy strategies were tested in the last years. For example, hypothermia treatment, neurotrophic factors or the blockage of excitotoxity. However, further studies are required to reveal the pressure independent pathomechanisms behind glaucoma. There is still an open issue whether immune mechanisms directly or indirectly trigger cell death pathways. Hence, it might be an imbalance between protective and destructive immune mechanisms. Moreover, identified therapy options have to be evaluated in more detail, since deeper insights could lead to better treatment options for glaucoma patients.
Collapse
Affiliation(s)
| | | | | | - Stephanie C. Joachim
- Experimental Eye Research, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| |
Collapse
|
30
|
Yang X, Zeng Q, Göktas E, Gopal K, Al-Aswad L, Blumberg DM, Cioffi GA, Liebmann JM, Tezel G. T-Lymphocyte Subset Distribution and Activity in Patients With Glaucoma. Invest Ophthalmol Vis Sci 2019; 60:877-888. [PMID: 30821813 PMCID: PMC6397017 DOI: 10.1167/iovs.18-26129] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Purpose Besides glia-driven neuroinflammation, growing evidence from analysis of human blood samples, isolated autoantibodies, and postmortem tissues also support systemic immune responses during neurodegeneration in glaucoma patients. To explore the T-cell–mediated component of systemic immunity, this study analyzed T lymphocytes in patients' blood. Methods Blood samples were collected from 32 patients with glaucoma and 21 nonglaucomatous controls, and mononuclear cells were isolated by Histopaque density gradient centrifugation. T-cell subset distribution was analyzed by multicolor flow cytometry after helper (Th) and cytotoxic fractions, and Th subpopulations, were stained with antibodies to CD4, CD8, or distinctive markers, such as IFN-γ (for Th1), IL-4 (for Th2), IL-17A (for Th17), and CD25/FoxP3 (for T regulatory cells [Tregs]). In addition, proliferative activity and cytokine secretion of T cells were analyzed after in vitro stimulation. Results Analysis of T-cell subset distribution detected a glaucoma-related shift. Despite similar frequencies of CD4+ or CD8+ T cells, or Th1, Th2, or Th17 subsets in glaucoma and control groups, glaucomatous samples exhibited a trend toward decreased frequency of CD4+ (or CD8+)/CD25+/FoxP3+ Tregs within the entire CD4+ (or CD8+) population (P < 0.001). Furthermore, CD4+ T cells in glaucomatous samples presented a greater stimulation response (∼3-fold) as characterized by increased proliferation and proinflammatory cytokine secretion (P < 0.05). Conclusions These findings suggest that the immunity activated in glaucoma may not be counterbalanced by an efficient immune suppression. More work is encouraged to determine whether shifted T-cell homeostasis may contribute to neurodegeneration in glaucoma, and/or whether T-cell subset imbalance may serve as a biomarker of autoimmune susceptibility.
Collapse
Affiliation(s)
- Xiangjun Yang
- Department of Ophthalmology, Columbia University, Vagelos College of Physicians and Surgeons, New York, New York, United States
| | - Qun Zeng
- Department of Ophthalmology, Columbia University, Vagelos College of Physicians and Surgeons, New York, New York, United States
| | - Emre Göktas
- Department of Ophthalmology, Columbia University, Vagelos College of Physicians and Surgeons, New York, New York, United States
| | - Kalashree Gopal
- Department of Ophthalmology, Columbia University, Vagelos College of Physicians and Surgeons, New York, New York, United States
| | - Lama Al-Aswad
- Department of Ophthalmology, Columbia University, Vagelos College of Physicians and Surgeons, New York, New York, United States
| | - Dana M Blumberg
- Department of Ophthalmology, Columbia University, Vagelos College of Physicians and Surgeons, New York, New York, United States
| | - George A Cioffi
- Department of Ophthalmology, Columbia University, Vagelos College of Physicians and Surgeons, New York, New York, United States
| | - Jeffrey M Liebmann
- Department of Ophthalmology, Columbia University, Vagelos College of Physicians and Surgeons, New York, New York, United States
| | - Gülgün Tezel
- Department of Ophthalmology, Columbia University, Vagelos College of Physicians and Surgeons, New York, New York, United States
| |
Collapse
|
31
|
Reinhard J, Wiemann S, Joachim SC, Palmhof M, Woestmann J, Denecke B, Wang Y, Downey GP, Faissner A. Heterozygous Meg2 Ablation Causes Intraocular Pressure Elevation and Progressive Glaucomatous Neurodegeneration. Mol Neurobiol 2019; 56:4322-4345. [PMID: 30315478 DOI: 10.1007/s12035-018-1376-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Accepted: 09/28/2018] [Indexed: 02/07/2023]
Abstract
Glaucomatous neurodegeneration represents one of the major causes of irreversible blindness worldwide. Yet, the detailed molecular mechanisms that initiate optic nerve damage and retinal ganglion cell (RGC) loss are not fully understood. Members of the protein tyrosine phosphatase (PTP) superfamily are key players in numerous neurodegenerative diseases. In order to investigate the potential functional relevance of the PTP megakaryocyte 2 (Meg2) in retinal neurodegeneration, we analyzed Meg2 knockout (KO) and heterozygous (HET)-synonym protein-tyrosine phosphatase non-receptor type 9 (Ptpn9)-mice. Interestingly, via global microarray and quantitative real-time PCR (RT-qPCR) analyses of Meg2 KO and HET retinae, we observed a dysregulation of several candidate genes that are highly associated with retinal degeneration and intraocular pressure (IOP) elevation, the main risk factor for glaucoma. Subsequent IOP measurements in Meg2 HET mice verified progressive age-dependent IOP elevation. Ultrastructural analyses and immunohistochemistry showed severe optic nerve degeneration accompanied by a dramatic loss of RGCs. Additionally, HET mice displayed reactive micro-/macrogliosis and early activation of the classical complement cascade with pronounced deposition of the membrane attack complex (MAC) in the retina and optic nerve. When treated with latanoprost, significant IOP lowering prevented RGC loss and microglial invasion in HET mice. Finally, electroretinogram (ERG) recordings revealed reduced a- and b-wave amplitudes, indicating impaired retinal functionality in Meg2 HET mice. Collectively, our findings indicate that the heterozygous loss of Meg2 in mice is sufficient to cause IOP elevation and glaucomatous neurodegeneration. Thus, Meg2 HET mice may serve as a novel animal model to study the pathomechanism involved in the onset and progression of glaucoma.
Collapse
Affiliation(s)
- Jacqueline Reinhard
- Department of Cell Morphology and Molecular Neurobiology, NDEF 05/594, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Universitätsstrasse 150, 44780, Bochum, Germany
| | - Susanne Wiemann
- Department of Cell Morphology and Molecular Neurobiology, NDEF 05/594, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Universitätsstrasse 150, 44780, Bochum, Germany
| | - Stephanie C Joachim
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, In der Schornau 23-25, 44892, Bochum, Germany
| | - Marina Palmhof
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, In der Schornau 23-25, 44892, Bochum, Germany
| | - Julia Woestmann
- Department of Cell Morphology and Molecular Neurobiology, NDEF 05/594, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Universitätsstrasse 150, 44780, Bochum, Germany
| | - Bernd Denecke
- Interdisciplinary Centre for Clinical Research, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Yingchun Wang
- Division of Respirology, Department of Medicine, University of Toronto and Toronto General Hospital Research Institute of the University Health Network, 610 University Avenue, Toronto, ON, M5S 1A8, Canada
| | - Gregory P Downey
- Division of Pulmonary Sciences and Critical Care Medicine, Departments of Medicine and Immunology and Microbiology, University of Colorado, Aurora, CO, 80045, USA
- Division of Pulmonary, Critical Care and Sleep Medicine, Departments of Medicine, Pediatrics and Biomedical Research, National Jewish Health, 1400 Jackson Street, Denver, CO, 80206, USA
| | - Andreas Faissner
- Department of Cell Morphology and Molecular Neurobiology, NDEF 05/594, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Universitätsstrasse 150, 44780, Bochum, Germany.
| |
Collapse
|
32
|
Zhao X, Kuang F, You YY, Wu MM, You SW. Etomidate affects the anti-oxidant pathway to protect retinal ganglion cells after optic nerve transection. Neural Regen Res 2019; 14:2020-2024. [PMID: 31290461 PMCID: PMC6676882 DOI: 10.4103/1673-5374.259627] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Our previous studies revealed that etomidate, a non-barbiturate intravenous anesthetic agent, has protective effects on retinal ganglion cells within 7 days after optic nerve transection. Whether this process is related to anti-oxidative stress is not clear. To reveal its mechanism, we established the optic nerve transection injury model by transecting 1 mm behind the left eyeball of adult male Sprague-Dawley rats. The rats received an intraperitoneal injection of etomidate (4 mg/kg) once per day for 7 days. The results showed that etomidate significantly enhanced the number of retinal ganglion cells retrogradely labeled with Fluorogold at 7 days after optic nerve transection. Etomidate also significantly reduced the levels of nitric oxide and malonaldehyde in the retina and increased the level of glutathione at 12 hours after optic nerve transection. Thus, etomidate can protect retinal ganglion cells after optic nerve transection in adult rats by activating an anti-oxidative stress response. The study was approved by the Animal Ethics Committee at Air Force Medical University, China (approval No. 20180305) on March 5, 2018.
Collapse
Affiliation(s)
- Xuan Zhao
- Department of Neurobiology and Collaborative Innovation Center for Brain Science, Air Force Medical University; Department of Histology and Embryology & Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical University, Xi'an, Shaanxi Province, China
| | - Fang Kuang
- Department of Neurobiology and Collaborative Innovation Center for Brain Science, Air Force Medical University, Xi'an, Shaanxi Province, China
| | - Yi-Yan You
- Ernest Mario School of Pharmacy, Rutgers, the State University of New Jersey, Piscataway, NJ, USA
| | - Ming-Mei Wu
- Department of Neurobiology and Collaborative Innovation Center for Brain Science, Air Force Medical University, Xi'an, Shaanxi Province, China
| | - Si-Wei You
- Department of Neurobiology and Collaborative Innovation Center for Brain Science, Air Force Medical University, Xi'an, Shaanxi Province, China
| |
Collapse
|
33
|
Abstract
Glaucoma is a chronic, progressive optic neuropathy characterized by the loss of peripheral vision first and then central vision. Clinically, normal tension glaucoma is considered a special subtype of glaucoma, in which the patient's intraocular pressure is within the normal range, but the patient experiences typical glaucomatous changes. However, increasing evidence has challenged the traditional pathophysiological view of normal tension glaucoma, which is based only on intraocular pressure, and breakthroughs in central nervous system imaging may now greatly increase our knowledge about the mechanisms underlying normal tension glaucoma. In this article, we review the latest progress in understanding the pathogenesis of normal tension glaucoma and in developing imaging techniques to detect it, to strengthen the appreciation for the connection between normal tension glaucoma and the brain.
Collapse
Affiliation(s)
- Hui-Jun Zhang
- Department of Ophthalmology, the First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, China
| | - Xue-Song Mi
- Department of Ophthalmology, the First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, China
| | - Kwok-Fai So
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, Guangdong Province; State Key Laboratory of Brain and Cognitive Sciences; Department of Ophthalmology, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| |
Collapse
|
34
|
Trivli A, Koliarakis I, Terzidou C, Goulielmos GN, Siganos CS, Spandidos DA, Dalianis G, Detorakis ET. Normal-tension glaucoma: Pathogenesis and genetics. Exp Ther Med 2018; 17:563-574. [PMID: 30651837 PMCID: PMC6307418 DOI: 10.3892/etm.2018.7011] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 11/21/2018] [Indexed: 12/27/2022] Open
Abstract
Normal-tension glaucoma (NTG) is a multifactorial optic neuropathy which, similar to open-angle glaucomas, is characterized by progressive retinal ganglion cell death and glaucomatous visual field loss. The major distinction of NTG from open-angle glaucomas is that the intraocular pressure (IOP) does not exceed the normal range. Missing the major risk factor and target of therapy, the elevated IOP, NTG poses a clinical challenge. Several insightful reviews have been published on the pathophysiology of NTG describing the possible underlying mechanisms. The current literature available also suggests that a significant percentage of patients with NTG (as high as 21%) have a family history of glaucoma, indicating a genetic predisposition to the disease. These facts strengthen the indication that NTG remains an enigmatic process. The aim of this review was to summarize the vascular, mechanical and genetic components considered to be responsible for NTG development and to discuss the mechanisms through which they are involved in the pathogenesis of NTG.
Collapse
Affiliation(s)
- Alexandra Trivli
- Department of Ophthalmology, Konstantopouleio-Patission General Hospital, 14233 Athens, Greece.,Section of Molecular Pathology and Human Genetics, Department of Internal Medicine, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Ioannis Koliarakis
- Laboratory of Anatomy-Histology-Embryology, Medical School, University of Crete, 71003 Heraklion, Greece
| | - Chryssa Terzidou
- Department of Ophthalmology, Konstantopouleio-Patission General Hospital, 14233 Athens, Greece
| | - George N Goulielmos
- Section of Molecular Pathology and Human Genetics, Department of Internal Medicine, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Charalambos S Siganos
- Department of Ophthalmology, University Hospital of Heraklion, 71110 Heraklion, Greece
| | - Demetrios A Spandidos
- Laboratory of Clinical Virology, Medical School, University of Crete, 71003 Heraklion, Greece
| | - Georgios Dalianis
- Department of Ophthalmology, Konstantopouleio-Patission General Hospital, 14233 Athens, Greece
| | | |
Collapse
|
35
|
Commensal microflora-induced T cell responses mediate progressive neurodegeneration in glaucoma. Nat Commun 2018; 9:3209. [PMID: 30097565 PMCID: PMC6086830 DOI: 10.1038/s41467-018-05681-9] [Citation(s) in RCA: 203] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 07/18/2018] [Indexed: 01/18/2023] Open
Abstract
Glaucoma is the most prevalent neurodegenerative disease and a leading cause of blindness worldwide. The mechanisms causing glaucomatous neurodegeneration are not fully understood. Here we show, using mice deficient in T and/or B cells and adoptive cell transfer, that transient elevation of intraocular pressure (IOP) is sufficient to induce T-cell infiltration into the retina. This T-cell infiltration leads to a prolonged phase of retinal ganglion cell degeneration that persists after IOP returns to a normal level. Heat shock proteins (HSP) are identified as target antigens of T-cell responses in glaucomatous mice and human glaucoma patients. Furthermore, retina-infiltrating T cells cross-react with human and bacterial HSPs; mice raised in the absence of commensal microflora do not develop glaucomatous T-cell responses or the associated neurodegeneration. These results provide compelling evidence that glaucomatous neurodegeneration is mediated in part by T cells that are pre-sensitized by exposure to commensal microflora.
Collapse
|
36
|
Adav SS, Wei J, Terence Y, Ang BCH, Yip LWL, Sze SK. Proteomic Analysis of Aqueous Humor from Primary Open Angle Glaucoma Patients on Drug Treatment Revealed Altered Complement Activation Cascade. J Proteome Res 2018; 17:2499-2510. [DOI: 10.1021/acs.jproteome.8b00244] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Sunil S. Adav
- School of Biological Sciences, Nanyang Technological University, Singapore 639798
| | - Jin Wei
- School of Biological Sciences, Nanyang Technological University, Singapore 639798
- Renmin Hospital of Wuhan University, Wuhan, Hubei 430072, PR China
| | - Yap Terence
- School of Biological Sciences, Nanyang Technological University, Singapore 639798
| | - Bryan C. H. Ang
- National Healthcare Group Eye Institute, Tan Tock Seng Hospital, Singapore 308433
| | - Leonard W. L. Yip
- National Healthcare Group Eye Institute, Tan Tock Seng Hospital, Singapore 308433
| | - Siu Kwan Sze
- School of Biological Sciences, Nanyang Technological University, Singapore 639798
| |
Collapse
|
37
|
Sharif NA. iDrugs and iDevices Discovery Research: Preclinical Assays, Techniques, and Animal Model Studies for Ocular Hypotensives and Neuroprotectants. J Ocul Pharmacol Ther 2018; 34:7-39. [PMID: 29323613 DOI: 10.1089/jop.2017.0125] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Discovery ophthalmic research is centered around delineating the molecular and cellular basis of ocular diseases and finding and exploiting molecular and genetic pathways associated with them. From such studies it is possible to determine suitable intervention points to address the disease process and hopefully to discover therapeutics to treat them. An investigational new drug (IND) filing for a new small-molecule drug, peptide, antibody, genetic treatment, or a device with global health authorities requires a number of preclinical studies to provide necessary safety and efficacy data. Specific regulatory elements needed for such IND-enabling studies are beyond the scope of this article. However, to enhance the overall data packages for such entities and permit high-quality foundation-building publications for medical affairs, additional research and development studies are always desirable. This review aims to provide examples of some target localization/verification, ocular drug discovery processes, and mechanistic and portfolio-enhancing exploratory investigations for candidate drugs and devices for the treatment of ocular hypertension and glaucomatous optic neuropathy (neurodegeneration of retinal ganglion cells and their axons). Examples of compound screening assays, use of various technologies and techniques, deployment of animal models, and data obtained from such studies are also presented.
Collapse
Affiliation(s)
- Najam A Sharif
- 1 Global Alliances & External Research , Santen Incorporated, Emeryville, California.,2 Department of Pharmaceutical Sciences, Texas Southern University , Houston, Texas.,3 Department of Pharmacology and Neuroscience, University of North Texas Health Sciences Center , Fort Worth, Texas
| |
Collapse
|
38
|
Reinhard J, Roll L, Faissner A. Tenascins in Retinal and Optic Nerve Neurodegeneration. Front Integr Neurosci 2017; 11:30. [PMID: 29109681 PMCID: PMC5660115 DOI: 10.3389/fnint.2017.00030] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 10/03/2017] [Indexed: 02/04/2023] Open
Abstract
Tenascins represent key constituents of the extracellular matrix (ECM) with major impact on central nervous system (CNS) development. In this regard, several studies indicate that they play a crucial role in axonal growth and guidance, synaptogenesis and boundary formation. These functions are not only important during development, but also for regeneration under several pathological conditions. Additionally, tenascin-C (Tnc) represents a key modulator of the immune system and inflammatory processes. In the present review article, we focus on the function of Tnc and tenascin-R (Tnr) in the diseased CNS, specifically after retinal and optic nerve damage and degeneration. We summarize the current view on both tenascins in diseases such as glaucoma, retinal ischemia, age-related macular degeneration (AMD) or diabetic retinopathy. In this context, we discuss their expression profile, possible functional relevance, remodeling of the interacting matrisome and tenascin receptors, especially under pathological conditions.
Collapse
Affiliation(s)
- Jacqueline Reinhard
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Bochum, Germany
| | - Lars Roll
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Bochum, Germany
| | - Andreas Faissner
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Bochum, Germany
| |
Collapse
|
39
|
Affiliation(s)
- Mohammadali Almasieh
- Departments of Ophthalmology and Neurology, McGill University, Montreal H4A 3S5, Canada
- Maisonneuve-Rosemont Hospital Research Center and Department of Ophthalmology, University of Montreal, Montreal H1T 2M4, Canada
| | - Leonard A. Levin
- Departments of Ophthalmology and Neurology, McGill University, Montreal H4A 3S5, Canada
- Maisonneuve-Rosemont Hospital Research Center and Department of Ophthalmology, University of Montreal, Montreal H1T 2M4, Canada
- Department of Ophthalmology and Visual Science, University of Wisconsin, Madison, Wisconsin 53706
| |
Collapse
|
40
|
Yang H, Reynaud J, Lockwood H, Williams G, Hardin C, Reyes L, Stowell C, Gardiner SK, Burgoyne CF. The connective tissue phenotype of glaucomatous cupping in the monkey eye - Clinical and research implications. Prog Retin Eye Res 2017; 59:1-52. [PMID: 28300644 PMCID: PMC5603293 DOI: 10.1016/j.preteyeres.2017.03.001] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 02/14/2017] [Accepted: 03/06/2017] [Indexed: 10/20/2022]
Abstract
In a series of previous publications we have proposed a framework for conceptualizing the optic nerve head (ONH) as a biomechanical structure. That framework proposes important roles for intraocular pressure (IOP), IOP-related stress and strain, cerebrospinal fluid pressure (CSFp), systemic and ocular determinants of blood flow, inflammation, auto-immunity, genetics, and other non-IOP related risk factors in the physiology of ONH aging and the pathophysiology of glaucomatous damage to the ONH. The present report summarizes 20 years of technique development and study results pertinent to the characterization of ONH connective tissue deformation and remodeling in the unilateral monkey experimental glaucoma (EG) model. In it we propose that the defining pathophysiology of a glaucomatous optic neuropathy involves deformation, remodeling, and mechanical failure of the ONH connective tissues. We view this as an active process, driven by astrocyte, microglial, fibroblast and oligodendrocyte mechanobiology. These cells, and the connective tissue phenomena they propagate, have primary and secondary effects on retinal ganglion cell (RGC) axon, laminar beam and retrolaminar capillary homeostasis that may initially be "protective" but eventually lead to RGC axonal injury, repair and/or cell death. The primary goal of this report is to summarize our 3D histomorphometric and optical coherence tomography (OCT)-based evidence for the early onset and progression of ONH connective tissue deformation and remodeling in monkey EG. A second goal is to explain the importance of including ONH connective tissue processes in characterizing the phenotype of a glaucomatous optic neuropathy in all species. A third goal is to summarize our current efforts to move from ONH morphology to the cell biology of connective tissue remodeling and axonal insult early in the disease. A final goal is to facilitate the translation of our findings and ideas into neuroprotective interventions that target these ONH phenomena for therapeutic effect.
Collapse
Affiliation(s)
- Hongli Yang
- Devers Eye Institute, Optic Nerve Head Research Laboratory, Legacy Research Institute, Portland, OR, United States; Devers Eye Institute, Discoveries in Sight Research Laboratories, Legacy Research Institute, Portland, OR, United States
| | - Juan Reynaud
- Devers Eye Institute, Optic Nerve Head Research Laboratory, Legacy Research Institute, Portland, OR, United States; Devers Eye Institute, Discoveries in Sight Research Laboratories, Legacy Research Institute, Portland, OR, United States
| | - Howard Lockwood
- Devers Eye Institute, Optic Nerve Head Research Laboratory, Legacy Research Institute, Portland, OR, United States; Devers Eye Institute, Discoveries in Sight Research Laboratories, Legacy Research Institute, Portland, OR, United States
| | - Galen Williams
- Devers Eye Institute, Optic Nerve Head Research Laboratory, Legacy Research Institute, Portland, OR, United States; Devers Eye Institute, Discoveries in Sight Research Laboratories, Legacy Research Institute, Portland, OR, United States
| | - Christy Hardin
- Devers Eye Institute, Optic Nerve Head Research Laboratory, Legacy Research Institute, Portland, OR, United States; Devers Eye Institute, Discoveries in Sight Research Laboratories, Legacy Research Institute, Portland, OR, United States
| | - Luke Reyes
- Devers Eye Institute, Optic Nerve Head Research Laboratory, Legacy Research Institute, Portland, OR, United States; Devers Eye Institute, Discoveries in Sight Research Laboratories, Legacy Research Institute, Portland, OR, United States
| | - Cheri Stowell
- Devers Eye Institute, Optic Nerve Head Research Laboratory, Legacy Research Institute, Portland, OR, United States; Devers Eye Institute, Discoveries in Sight Research Laboratories, Legacy Research Institute, Portland, OR, United States
| | - Stuart K Gardiner
- Devers Eye Institute, Discoveries in Sight Research Laboratories, Legacy Research Institute, Portland, OR, United States
| | - Claude F Burgoyne
- Devers Eye Institute, Optic Nerve Head Research Laboratory, Legacy Research Institute, Portland, OR, United States; Devers Eye Institute, Discoveries in Sight Research Laboratories, Legacy Research Institute, Portland, OR, United States.
| |
Collapse
|
41
|
Tong Y, Zhou YL, Zheng Y, Biswal M, Zhao PQ, Wang ZY. Analyzing cytokines as biomarkers to evaluate severity of glaucoma. Int J Ophthalmol 2017; 10:925-930. [PMID: 28730084 DOI: 10.18240/ijo.2017.06.15] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 02/06/2017] [Indexed: 11/23/2022] Open
Abstract
AIM To analyze cytokines as biomarkers for evaluation of severity of glaucoma. METHODS This was a prospective case-control study including 29 eyes with glaucoma. Besides, 28 eyes with senile cataract were used as control. Patients were classified into four groups: acute angle closure glaucoma (AACG), chronic angle closure glaucoma (CACG), primary open angle glaucoma (POAG) and senile cataract. Undiluted vitreous samples were collected, then vitreous concentrations of 9 types of cytokines were determined by cytometric bead assay system: γ-interferon (IFNg), interleukin (IL)-10, IL-2, IL-4, IL-5, interferon-γ-inducible protein (IP)-10, monocyte chemoattractant protein (MCP)-1, tumor necrosis factor (TNF)-α, and vascular endothelial growth factor (VEGF). We also recorded the intraocular pressure (IOP) of patients in each group and Pearson correlated analysis was performed to analysis the correlation between each type of cytokine with IOP. RESULTS Vitreous levels of IL-2, IL-5, MCP-1, TNF-α and IP-10 were significantly higher (P<0.05) in AACG group. Patients with AACG, CACG and POAG have higher IOP than senile cataract, but we didn't find any significant correlation between IOP with any type of the cytokines. CONCLUSION Inflammation and immune reaction have a strong link with the pathology of glaucoma especially AACG. Some cytokines may act as biomarkers to evaluate the severity of glaucoma. Anti-inflammatory treatments and controlling of IOP are necessary for the therapy of glaucoma.
Collapse
Affiliation(s)
- Yao Tong
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200011, China.,Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Ya-Li Zhou
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200011, China
| | - Yan Zheng
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, China.,Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Chongming Branch, Shanghai 202150, China
| | - Manas Biswal
- Department of Molecular Genetics, University of Florida, Gainesville, Florida 32610, USA
| | - Pei-Quan Zhao
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Zhao-Yang Wang
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200011, China
| |
Collapse
|
42
|
Williams PA, Marsh-Armstrong N, Howell GR. Neuroinflammation in glaucoma: A new opportunity. Exp Eye Res 2017; 157:20-27. [PMID: 28242160 PMCID: PMC5497582 DOI: 10.1016/j.exer.2017.02.014] [Citation(s) in RCA: 185] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 02/16/2017] [Accepted: 02/23/2017] [Indexed: 10/20/2022]
Abstract
Mounting evidence suggests neuroinflammation is a key process in glaucoma, yet the precise roles are not known. Understanding these complex processes, which may also be a key in other common neurodegenerations such as Alzheimer's disease, will lead to targeted therapeutics for a disease that affects as many as 80 million people worldwide. Here, we define neuroinflammation as any immune-relevant response by a variety of cell types including astrocytes, microglia, and peripherally derived cells occurring in the optic nerve head and/or retina. In this review article, we first discuss clinical evidence for neuroinflammation in glaucoma and define neuroinflammation in glaucoma. We then review the inflammatory pathways that have been associated with glaucoma. Finally, we set out key research directions that we believe will greatly advance our understanding of the role of neuroinflammation in glaucoma. This review arose from a discussion of neuroinflammation in glaucoma at the 2015 meeting of The Lasker/IRRF Initiative for Innovation in Vision Science. This manuscript sets out to summarize one of these sessions; "Inflammation and Glaucomatous Neurodegeneration", as well as to review the current state of the literature surrounding neuroinflammation in glaucoma.
Collapse
Affiliation(s)
| | - Nick Marsh-Armstrong
- The Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; Hugo W. Moser Research Institute, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Gareth R Howell
- The Jackson Laboratory, Bar Harbor, ME, USA; Graduate Program of Genetics, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, MA, USA.
| |
Collapse
|
43
|
Khalef N, Labib H, Helmy H, El Hamid MA, Moemen L, Fahmy I. Levels of cytokines in the aqueous humor of eyes with primary open angle glaucoma, pseudoexfoliation glaucoma and cataract. Electron Physician 2017; 9:3833-3837. [PMID: 28465815 PMCID: PMC5410914 DOI: 10.19082/3833] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2016] [Accepted: 01/16/2017] [Indexed: 12/01/2022] Open
Abstract
Objective The focus of this study aimed at measuring multiple inflammatory cytokines levels in the aqueous humor of patients with primary open angle glaucoma (POAG), pseudoexfoliation glaucoma (PEXG) and senile cataract. Methods This case control study was conducted at the Research Institute of Ophthalmology, Giza, Egypt in 2016. Aqueous humor (AH) samples were withdrawn from 50 patients (30 POAG, and 20 PEXG) and from 15 patients with senile cataract serving as controls. The levels of IL6, IL8, transforming growth factor β1 (TGFβ1), tumor necrosis growth factor α (TNFα) and serum amyloid A (SAA) were analyzed by ELISA immune-assay. Data were analyzed by SPSS 10, using Pearson Product-Moment Correlation and independent-samples t-test. Results The levels of IL8, TGFβ1, TNFα and SAA were significantly higher in POAG and PEXG patients, compared to senile cataract patients. While the levels of IL6, were significantly decreased in both groups of glaucoma patients compared to cataract patients. Significant positive correlations were detected between IL6, IL 8 & TGF β1, IL 8; SAA, IL8 & TGFβ1, SAA in the aqueous humor of different groups. Conclusion Thus the assayed cytokines including TGFβ1, TNFα, IL8 and SAA in aqueous humor, play a vital role in IOP elevations in patients with POAG and PEXG.
Collapse
Affiliation(s)
- Nervana Khalef
- Clinical Pathology Unit, Research Institute of Ophthalmology, Giza, Egypt
| | - Hany Labib
- Ophthalmology Department, Research Institute of Ophthalmology, Giza, Egypt
| | - Hazem Helmy
- Ophthalmology Department, Research Institute of Ophthalmology, Giza, Egypt
| | - Mona Abd El Hamid
- Medical Biochemistry Unit, Research Institute of Ophthalmology, Giza, Egypt
| | - Leqaa Moemen
- Medical Biochemistry Unit, Research Institute of Ophthalmology, Giza, Egypt
| | - Iman Fahmy
- Ophthalmology Department, Research Institute of Ophthalmology, Giza, Egypt
| |
Collapse
|
44
|
Lim JC, Lu W, Beckel JM, Mitchell CH. Neuronal Release of Cytokine IL-3 Triggered by Mechanosensitive Autostimulation of the P2X7 Receptor Is Neuroprotective. Front Cell Neurosci 2016; 10:270. [PMID: 27932954 PMCID: PMC5120082 DOI: 10.3389/fncel.2016.00270] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 11/07/2016] [Indexed: 12/05/2022] Open
Abstract
Mechanical strain due to increased pressure or swelling activates inflammatory responses in many neural systems. As cytokines and chemokine messengers lead to both pro-inflammatory and neuroprotective actions, understanding the signaling patterns triggered by mechanical stress may help improve overall outcomes. While cytokine signaling in neural systems is often associated with glial cells like astrocytes and microglia, the contribution of neurons themselves to the cytokine response is underappreciated and has bearing on any balanced response. Mechanical stretch of isolated neurons was previously shown to trigger ATP release through pannexin hemichannels and autostimulation of P2X7 receptors (P2X7Rs) on the neural membrane. Given that P2X7Rs are linked to cytokine activation in other cells, this study investigates the link between neuronal stretch and cytokine release through a P2X7-dependent pathway. Cytokine assays showed application of a 4% strain to isolated rat retinal ganglion cells (RGCs) released multiple cytokines. The P2X7R agonist BzATP also released multiple cytokines; Interleukin 3 (IL-3), TNF-α, CXCL9, VEGF, L-selectin, IL-4, GM-CSF, IL-10, IL-1Rα, MIP and CCL20 were released by both stimuli, with the release of IL-3 greatest with either stimuli. Stretch-dependent IL-3 release was confirmed with ELISA and blocked by P2X7R antagonists A438079 and Brilliant Blue G (BBG), implicating autostimulation of the P2X7R in stretch-dependent IL-3 release. Neuronal IL-3 release triggered by BzATP required extracellular calcium. The IL-3Rα receptor was expressed on RGCs but not astrocytes, and both IL-3Rα and IL-3 itself were predominantly expressed in the retinal ganglion cell layer of adult retinal sections, implying autostimulation of receptors by released IL-3. While the number of surviving ganglion cells decreased with time in culture, the addition of IL-3 protected against this loss of neurons. Expression of mRNA for IL-3 and IL-3Rα increased in rat retinas stretched with moderate intraocular pressure (IOP) elevation; BBG blocked the rise in IL-3, implicating a role for the P2X7R in transcriptional regulation in vivo. In summary, mechanical stretch triggers release of cytokines from neurons that can convey neuroprotection. The enhancement of these signals in vivo implicates P2X7R-mediated IL-3 signaling as an endogenous pathway that could minimize damage following neuronal exposure to chronic mechanical strain.
Collapse
Affiliation(s)
- Jason C Lim
- Department of Anatomy and Cell Biology, University of Pennsylvania Philadelphia, PA, USA
| | - Wennan Lu
- Department of Anatomy and Cell Biology, University of Pennsylvania Philadelphia, PA, USA
| | - Jonathan M Beckel
- Department of Pharmacology and Chemical Biology, University of Pittsburgh Pittsburgh, PA, USA
| | - Claire H Mitchell
- Department of Anatomy and Cell Biology, University of PennsylvaniaPhiladelphia, PA, USA; Department of Physiology, University of PennsylvaniaPhiladelphia, PA, USA; Department of Ophthalmology, University of PennsylvaniaPhiladelphia, PA, USA
| |
Collapse
|
45
|
Moazzeni H, Akbari MT, Yazdani S, Elahi E. Expression of CXCL6 and BBS5 that may be glaucoma relevant genes is regulated by PITX2. Gene 2016; 593:76-83. [PMID: 27520585 DOI: 10.1016/j.gene.2016.08.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 07/31/2016] [Accepted: 08/08/2016] [Indexed: 12/26/2022]
Abstract
The transcription factor PITX2 is implicated in glaucoma pathology. In an earlier study we had used microarray analysis to identify genes in the trabecular meshwork (TM) that are affected by knock down of PITX2. Here, those studies were pursued to identify genes that are direct targets of PITX2 and that may be relevant to glaucoma. Initially, bioinformatics tools were used to select among the genes that had been affected by PITX2 knock down those that have PITX2 binding sites and that may be involved in glaucoma related functions. Subsequently, the effect of PITX2 was tested using the dual luciferase assay in four cell cultures including two primary TM cultures co-transfected with vectors containing promoter fragments of six candidate genes upstream of a luciferase gene and a vector that expressed PITX2. Finally, the effect of PITX2 on endogenous expression of two genes was assessed by over expression and knock down of PITX2 in TM cells. Thirty four genes were found to contain PITX2 binding sites in their putative promoter regions, and 16 were found to be associated with TM-specific and/or glaucoma associated functions. Results of dual luciferase assays confirmed that two of six genes tested were directly targeted by PITX2. The two genes were CXCL6 (chemokine (C-X-C motif) ligand 6) and BBS5 (Bardet-Biedl syndrome 5). Over expression and knock down of PITX2 showed that this transcription factor affects endogenous expression of these two genes in TM cells. CXCL6 encodes a pro-inflammatory cytokine, and many studies have suggested that cytokines and other immune system functions are involved in glaucoma pathogenesis. BBS5 is a member of the BBS family of genes that affect ciliary functions, and ciliary bodies in the anterior chamber of the eye produce the aqueous fluid that affects intraocular pressure. Immune related functions and intraocular pressure are both important components of glaucoma pathology. The role of PITX2 in glaucoma may be mediated partly by regulating the expression of CXCL6 and BBS5 and thus affecting immune functions and intraocular pressure.
Collapse
Affiliation(s)
- Hamidreza Moazzeni
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, P.O. Box. 14115-331, Tehran, Iran
| | - Mohammad Taghi Akbari
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, P.O. Box. 14115-331, Tehran, Iran.
| | - Shahin Yazdani
- Ophthalmic Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Elahe Elahi
- School of Biology, College of Science, University of Tehran, Tehran, Iran; Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran.
| |
Collapse
|
46
|
Weinreb RN, Leung CKS, Crowston JG, Medeiros FA, Friedman DS, Wiggs JL, Martin KR. Primary open-angle glaucoma. Nat Rev Dis Primers 2016; 2:16067. [PMID: 27654570 DOI: 10.1038/nrdp.2016.67] [Citation(s) in RCA: 331] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Glaucoma is an optic neuropathy that is characterized by the progressive degeneration of the optic nerve, leading to visual impairment. Glaucoma is the main cause of irreversible blindness worldwide, but typically remains asymptomatic until very severe. Open-angle glaucoma comprises the majority of cases in the United States and western Europe, of which, primary open-angle glaucoma (POAG) is the most common type. By contrast, in China and other Asian countries, angle-closure glaucoma is highly prevalent. These two types of glaucoma are characterized based on the anatomic configuration of the aqueous humour outflow pathway. The pathophysiology of POAG is not well understood, but it is an optic neuropathy that is thought to be associated with intraocular pressure (IOP)-related damage to the optic nerve head and resultant loss of retinal ganglion cells (RGCs). POAG is generally diagnosed during routine eye examination, which includes fundoscopic evaluation and visual field assessment (using perimetry). An increase in IOP, measured by tonometry, is not essential for diagnosis. Management of POAG includes topical drug therapies and surgery to reduce IOP, although new therapies targeting neuroprotection of RGCs and axonal regeneration are under development.
Collapse
Affiliation(s)
- Robert N Weinreb
- Shiley Eye Institute, Hamilton Glaucoma Center, Department of Ophthalmology, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, USA
| | - Christopher K S Leung
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Jonathan G Crowston
- Department of Ophthalmology, Centre for Eye Research Australia, University of Melbourne, Melbourne, Victoria, Australia
| | - Felipe A Medeiros
- Shiley Eye Institute, Hamilton Glaucoma Center, Department of Ophthalmology, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, USA
| | - David S Friedman
- Dana Center for Preventive Ophthalmology, Johns Hopkins Wilmer Eye Institute, Baltimore, Maryland, USA
| | - Janey L Wiggs
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, USA
| | - Keith R Martin
- Department of Ophthalmology and Cambridge NIHR Biomedical Research Centre, University of Cambridge, Cambridge, UK
| |
Collapse
|
47
|
Mookherjee S, Banerjee D, Chakraborty S, Mukhopadhyay I, Sen A, Ray K. Evaluation of the IL1 Gene Cluster Single Nucleotide Polymorphisms in Primary Open-Angle Glaucoma Pathogenesis. Genet Test Mol Biomarkers 2016; 20:633-636. [PMID: 27533638 DOI: 10.1089/gtmb.2015.0344] [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] [Indexed: 01/28/2023] Open
Abstract
AIMS Dysregulation of the immune system has previously been implicated in glaucoma pathogenesis. In this study, we investigated the potential association of SNPs in the IL1 gene cluster, consisting of nine genes, with primary open-angle glaucoma (POAG) cases. These cases presented with low to normal intraocular pressures (<20 mmHg), and are referred to as non-high tension glaucoma (non-HTG) cases. MATERIALS AND METHODS In this biphasic study, the discovery phase was conducted with 198 non-HTG cases and 112 controls from eastern India. A total of 68 single nucleotide polymorphisms (SNPs) spanning the IL1 nine-gene cluster region were genotyped using the MALDI-TOF based Sequenom platform. SNPs, which were found to be significantly associated with non-HTG cases in the first phase of the study, were further genotyped by Sanger sequencing in a replication cohort consisting of 194 non-HTG cases and 242 controls. RESULTS In the discovery phase, two nonsynonymous SNPs (rs3811046 and rs3811047), located in the IL1F7 gene and in an intergenic region, respectively were found to be weakly associated with non-HTG cases. However, the association was not sustained in the replication cohort. CONCLUSION Our study did not reveal any reproducible association of SNPs in the IL1 gene cluster with POAG.
Collapse
Affiliation(s)
- Suddhasil Mookherjee
- 1 Molecular & Human Genetics Division, CSIR-Indian Institute of Chemical Biology , Kolkata, India
| | - Deblina Banerjee
- 1 Molecular & Human Genetics Division, CSIR-Indian Institute of Chemical Biology , Kolkata, India
| | - Subhadip Chakraborty
- 1 Molecular & Human Genetics Division, CSIR-Indian Institute of Chemical Biology , Kolkata, India
| | | | | | - Kunal Ray
- 1 Molecular & Human Genetics Division, CSIR-Indian Institute of Chemical Biology , Kolkata, India
| |
Collapse
|
48
|
Reinehr S, Reinhard J, Wiemann S, Stute G, Kuehn S, Woestmann J, Dick HB, Faissner A, Joachim SC. Early remodelling of the extracellular matrix proteins tenascin-C and phosphacan in retina and optic nerve of an experimental autoimmune glaucoma model. J Cell Mol Med 2016; 20:2122-2137. [PMID: 27374750 PMCID: PMC5082392 DOI: 10.1111/jcmm.12909] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 04/19/2016] [Indexed: 12/17/2022] Open
Abstract
Glaucoma is characterized by the loss of retinal ganglion cells (RGCs) and optic nerve fibres. Previous studies noted fewer RGCs after immunization with ocular antigens at 28 days. It is known that changes in extracellular matrix (ECM) components conduct retina and optic nerve degeneration. Here, we focused on the remodelling of tenascin‐C and phosphacan/receptor protein tyrosine phosphatase β/ζ in an autoimmune glaucoma model. Rats were immunized with optic nerve homogenate (ONA) or S100B protein (S100). Controls received sodium chloride (Co). After 14 days, no changes in RGC number were noted in all groups. An increase in GFAPmRNA expression was observed in the S100 group, whereas no alterations were noted via immunohistochemistry in both groups. Extracellular matrix remodelling was analyzed after 3, 7, 14 and 28 days. Tenascin‐C and 473HD immunoreactivity in retinae and optic nerves was unaltered in both immunized groups at 3 days. At 7 days, tenascin‐C staining increased in both tissues in the ONA group. Also, in the optic nerves of the S100 group, an intense tenascin‐C staining could be shown. In the retina, an increased tenascin‐C expression was also observed in ONA animals via Western blot. 473HD immunoreactivity was elevated in the ONA group in both tissues and in the S100 optic nerves at 7 days. At 14 days, tenascin‐C and 473HD immunoreactivity was up‐regulated in the ONA retinae, whereas phosphacan expression was up‐regulated in both groups. We conclude that remodelling of tenascin‐C and phosphacan occurred shortly after immunization, already before RGC loss. We assume that both ECM molecules represent early indicators of neurodegeneration.
Collapse
Affiliation(s)
- Sabrina Reinehr
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Jacqueline Reinhard
- Department of Cell Morphology and Molecular Neurobiology, Ruhr-University Bochum, Bochum, Germany
| | - Susanne Wiemann
- Department of Cell Morphology and Molecular Neurobiology, Ruhr-University Bochum, Bochum, Germany
| | - Gesa Stute
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Sandra Kuehn
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Julia Woestmann
- Department of Cell Morphology and Molecular Neurobiology, Ruhr-University Bochum, Bochum, Germany
| | - H Burkhard Dick
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Andreas Faissner
- Department of Cell Morphology and Molecular Neurobiology, Ruhr-University Bochum, Bochum, Germany.
| | - Stephanie C Joachim
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany.
| |
Collapse
|
49
|
Kumarasamy N, Lam F, Wang A, Theoharides T. Glaucoma: Current and Developing Concepts for Inflammation, Pathogenesis and Treatment. EUR J INFLAMM 2016. [DOI: 10.1177/1721727x0600400301] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Glaucoma is a prevalent neurodegenerative disorder of the eye. However, the mechanism leading to the disease is still unclear. Increased intraocular pressure (IOP) and subsequent retinal ganglion cell (RGC) death leading to the loss of visual field characterizes the pathology of primary open angle glaucoma, which is the most common form. Possible factors leading to glaucoma include glutamate-induced neurotoxicity, nitric oxide (NO) based damage, disruption of neurotrophic factor transport and immune-induced neurodestruction. Current treatment options primarily aim at decreasing IOP by utilizing pharmacological agents, laser therapy and surgery. Developing treatments target neuroprotection with vaccines, the inhibition of NO synthesis and apoptosis. Gaining a better understanding of the pathogenesis can aid in the development of new treatment options and, perhaps, even a cure for glaucoma.
Collapse
Affiliation(s)
| | | | | | - T.C. Theoharides
- Departments of Pharmacology and Experimental Therapeutics, Internal Medicine and Biochemistry, Tufts University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
50
|
Gramlich OW, Teister J, Neumann M, Tao X, Beck S, von Pein HD, Pfeiffer N, Grus FH. Immune response after intermittent minimally invasive intraocular pressure elevations in an experimental animal model of glaucoma. J Neuroinflammation 2016; 13:82. [PMID: 27090083 PMCID: PMC4836145 DOI: 10.1186/s12974-016-0542-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 04/07/2016] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Elevated intraocular pressure (IOP), as well as fluctuations in IOP, is a main risk factor for glaucoma, but its pathogenic effect has not yet been clarified. Beyond the multifactorial pathology of the disease, autoimmune mechanisms seem to be linked to retinal ganglion cell (RGC) death. This study aimed to identify if intermittent IOP elevations in vivo (i) elicit neurodegeneration, (ii) provokes an immune response and (iii) whether progression of RGC loss can be attenuated by the B lymphocyte inhibitor Belimumab. METHODS Using an intermittent ocular hypertension model (iOHT), Long Evans rats (n = 21) underwent 27 unilateral simulations of a fluctuating pressure profile. Nine of these animals received Belimumab, and additional seven rats served as normotensive controls. Axonal density was analyzed in PPD-stained optic nerve cross-sections. Retinal cross-sections were immunostained against Brn3a, Iba1, and IgG autoantibody depositions. Serum IgG concentration and IgG reactivities were determined using ELISA and protein microarrays. Data was analyzed using ANOVA and Tukey HSD test (unequal N) or student's independent t test by groups. RESULTS A wavelike IOP profile led to a significant neurodegeneration of optic nerve axons (-10.6 %, p < 0.001) and RGC (-19.5 %, p = 0.02) in iOHT eyes compared with fellow eyes. Belimumab-treated animals only showed slightly higher axonal survival and reduced serum IgG concentration (-29 %) after iOHT. Neuroinflammatory events, indicated by significantly upregulated microglia activation and IgG autoantibody depositions, were shown in all injured retinas. Significantly elevated serum autoantibody immunoreactivities against glutathione-S-transferase, spectrin, and transferrin were observed after iOHT and were negatively correlated to the axon density. CONCLUSIONS Intermittent IOP elevations are sufficient to provoke neurodegeneration in the optic nerve and the retina and elicit changes of IgG autoantibody reactivities. Although the inhibition of B lymphocyte activation failed to ameliorate axonal survival, the correlation between damage and changes in the autoantibody reactivity suggests that autoantibody profiling could be useful as a biomarker for glaucoma.
Collapse
Affiliation(s)
- Oliver W Gramlich
- Experimental Ophthalmology, Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany.,Glaucoma Cell Biology Laboratory, Department of Ophthalmology and Visual Sciences, The University of Iowa, Iowa City, IA, 62242, USA
| | - Julia Teister
- Experimental Ophthalmology, Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Mareike Neumann
- Experimental Ophthalmology, Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Xue Tao
- Experimental Ophthalmology, Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Sabine Beck
- Experimental Ophthalmology, Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Harald D von Pein
- Department of Neuropathology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, Mainz, 55131, Germany
| | - Norbert Pfeiffer
- Experimental Ophthalmology, Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Franz H Grus
- Experimental Ophthalmology, Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany.
| |
Collapse
|