1
|
Li B, Zhao C, Guo S, Li X, Zhang H, Duan Y, Zhang M, Tao Q, Zhou P, Li X, Zhang X. Recombinant adeno-associated virus with anti-tumor necrosis factor-alpha in an experimental autoimmune uveitis model. Exp Eye Res 2025; 253:110273. [PMID: 39922525 DOI: 10.1016/j.exer.2025.110273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 01/29/2025] [Accepted: 02/05/2025] [Indexed: 02/10/2025]
Abstract
Uveitis treatment is associated with side effects and inconsistent outcomes. Existing treatments often fail to provide targeted and sustained relief; thus, novel therapeutic approaches are needed. Among these, gene therapy using adeno-associated virus (AAV) vectors target specific retinal cells, show low immunogenicity, and demonstrate sustained gene expression, making it a potential advancement in uveitis treatment. Therefore, we utilized a AAV2 system encapsulating encoded anti-tumor necrosis factor-alpha (TNF-α) antibody to assess its efficacy in the treatment of experimental autoimmune uveitis (EAU) in mice. Compared with the AAV2-GFP group, AAV2-ADA-injected mice showed significantly reduced clinical, OCT, and histopathological scores in EAU with lower percentages of Th1 and Th17 cells in the eyes and higher percentages of Treg cells in the draining lymph nodes (LN). This study demonstrated the safety and effects of AAV2-ADA in EAU treatment, providing a promising therapeutic strategy for uveitis.
Collapse
Affiliation(s)
- Baiyi Li
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China
| | - Chuan Zhao
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China
| | | | - Xueru Li
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China
| | - Hui Zhang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China
| | - Yanan Duan
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China
| | - Mi Zhang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China
| | - Qingqin Tao
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China
| | - Peiran Zhou
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China
| | - Xiaorong Li
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China
| | - Xiaomin Zhang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China.
| |
Collapse
|
2
|
Yuan M, Yi S, Wang X, Han G, Wei P, Lv Z, Gui B, Chen X, Wang Y, Zhu L. Promoted Translocation of Perfluorooctanoic Acid across the Blood-Retinal Barrier due to its Inhibition of Tight Junction Assembly by Antagonizing LPAR1. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2025; 59:4807-4819. [PMID: 40038073 DOI: 10.1021/acs.est.4c12051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
Abstract
Eye health is becoming a significant public health concern, and a recent epidemiological investigation suggested that perfluorooctanoic acid (PFOA), a so-called forever chemical, was correlated with decreased human visual acuity; however, it remains unknown whether PFOA can pass through the blood-retinal barrier (BRB) to cause visual toxicity. In this study, the mice received a 28-day subchronic oral exposure to PFOA. The results of spatial mass spectrometry imaging indicated that the eye-enriched PFOA dispersed into the subretina primarily through the outer BRB (oBRB), which subsequently resulted in significantly increased apoptosis and decreased thickness of multiple oBRB-associated layers. BRB integrity and function were compromised due to decreased expression of the tight junction (TJ). Mechanistically, PFOA outcompeted lysophosphatidic acid to bind strongly with lysophosphatidic acid receptor 1 (LPAR1) in its antagonism, abolishing its ability to stimulate the TJ assembly-related signaling pathway. This subsequently attenuated phosphorylation of the myosin light chain, rendering insufficient contraction of the actomyosin cytoskeleton, leading to decreased TJ assembly and BRB leakage. This, in turn, facilitated PFOA translocation across the BRB and accumulation within the subretinal space. Our findings suggest that oBRB is particularly vulnerable to PFOA, which targets directly LPAR1 to disable its function of maintaining TJ assembly cascades, leading to adverse visual effects.
Collapse
Affiliation(s)
- Meng Yuan
- Key Laboratory of Pollution Processes and Environmental Criteria of Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering, Nankai University, Tianjin 300350, China
| | - Shujun Yi
- Key Laboratory of Pollution Processes and Environmental Criteria of Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering, Nankai University, Tianjin 300350, China
| | - Xiaoyan Wang
- Key Laboratory of Pollution Processes and Environmental Criteria of Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering, Nankai University, Tianjin 300350, China
| | - Guoge Han
- Tianjin Eye Hospital, Tianjin Key Laboratory of Ophthalmology and Visual Science, Nankai University Eye Institute, Nankai University Affiliated Eye Hospital, Nankai University, Tianjin 300020, China
| | - Pinghui Wei
- Tianjin Eye Hospital, Tianjin Key Laboratory of Ophthalmology and Visual Science, Nankai University Eye Institute, Nankai University Affiliated Eye Hospital, Nankai University, Tianjin 300020, China
| | - Zixuan Lv
- Key Laboratory of Pollution Processes and Environmental Criteria of Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering, Nankai University, Tianjin 300350, China
| | - Bingxin Gui
- Key Laboratory of Pollution Processes and Environmental Criteria of Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering, Nankai University, Tianjin 300350, China
| | - Xuan Chen
- Tianjin Eye Hospital, Tianjin Key Laboratory of Ophthalmology and Visual Science, Nankai University Eye Institute, Nankai University Affiliated Eye Hospital, Nankai University, Tianjin 300020, China
| | - Yan Wang
- Tianjin Eye Hospital, Tianjin Key Laboratory of Ophthalmology and Visual Science, Nankai University Eye Institute, Nankai University Affiliated Eye Hospital, Nankai University, Tianjin 300020, China
| | - Lingyan Zhu
- Key Laboratory of Pollution Processes and Environmental Criteria of Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering, Nankai University, Tianjin 300350, China
| |
Collapse
|
3
|
Chenjin Z. Exposure assessments of cadmium and lead with age-related eye disease: A systematic review and meta-analysis. J Trace Elem Med Biol 2025; 89:127631. [PMID: 40122018 DOI: 10.1016/j.jtemb.2025.127631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 02/21/2025] [Accepted: 03/06/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Age-related eye diseases, such as macular degeneration, cataracts, and glaucoma, are significant causes of vision loss in older adults. Emerging evidence suggests that environmental exposure to heavy metals, particularly Cadmium and lead, may play an essential role in the onset and progression of these conditions. METHODS This systematic review and meta-analysis aimed to evaluate the association between Cadmium and lead exposure and the risk of age-related eye diseases. A comprehensive literature search was conducted across multiple databases, including PubMed, Embase, Scopus, and Web of Science, covering studies published up to July 2024. The review included observational studies on the relationship between Cadmium or lead exposure and specific eye conditions. Data extraction and quality assessment were performed independently by two reviewers. The meta-analysis utilized a random effects model to analyze cadmium and lead levels in patient and control groups, with subgroup analyses based on the type of eye disease and study design. RESULTS The results revealed a significant association between cadmium exposure and an increased risk of eye diseases, particularly cataracts and glaucoma. Lead exposure was also linked to a higher risk of cataracts and macular degeneration. The study found substantial heterogeneity among the included studies, highlighting the variability in exposure assessment and population characteristics. CONCLUSIONS Despite these variations, the findings underscore the importance of addressing environmental exposures to toxic metals as potential risk factors for age-related eye diseases. Further research is needed to clarify the underlying mechanisms and to develop targeted interventions for reducing these risks.
Collapse
Affiliation(s)
- Zhao Chenjin
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610000, China.
| |
Collapse
|
4
|
Khidr EG, Morad NI, Hatem S, El-Dessouki AM, Mohamed AF, El-Shiekh RA, Hafeez MSAE, Ghaiad HR. Natural remedies proposed for the management of diabetic retinopathy (DR): diabetic complications. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03866-w. [PMID: 39954069 DOI: 10.1007/s00210-025-03866-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 01/28/2025] [Indexed: 02/17/2025]
Abstract
Diabetic retinopathy (DR) represents a significant and serious complication associated with diabetes mellitus (DM), often resulting in considerable visual impairment or even blindness. The intricate pathological processes underlying DR complicate the effectiveness of current treatment modalities. Studies have highlighted the potential of natural products in the treatment of DR via several beneficial effects including anti-inflammatory, antioxidant, anti-neovascular, and anti-apoptotic properties. Flavonoids, saponins, saccharides, and alkaloids exhibited various beneficial effects in DR in in vivo and in vitro studies. However, the clinical utilization of these natural compounds is hindered by issues such as inadequate specificity, low bioavailability, and potential toxicity. Therefore, there is a pressing need for rigorous clinical studies to confirm the efficacy of natural products in preventing or mitigating the progression of DR.
Collapse
Affiliation(s)
- Emad Gamil Khidr
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| | - Nourhan Ibrahim Morad
- Department of Pharmacognosy and Natural Products, Faculty of Pharmacy, Menofia University, Menofia, Egypt
| | - Shymaa Hatem
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Future University in Egypt, Cairo, Egypt.
| | - Ahmed M El-Dessouki
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Ahram Canadian University, 6Th of October City, Giza, 12566, Egypt
| | - Ahmed F Mohamed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
- Faculty of Pharmacy, King Salman International University (KSIU), Ras Sedr, South Sinai, 46612, Egypt
| | - Riham A El-Shiekh
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo, 11562, Egypt.
| | - Mohamed S Abd El Hafeez
- Department of Pharmacy, Kut University College, Al Kut, Wasit, 52001, Iraq
- Department of Pharmacognosy, Faculty of Pharmacy, Egyptian Russian University, Cairo-Suez Road, Badr, 11829, Egypt
| | - Heba R Ghaiad
- Biochemistry Department, Faculty of Pharmacy, Cairo University, Kasr El Ainy St., Cairo, 11562, Egypt
| |
Collapse
|
5
|
Shah W, Gong Y, Qiao X, Lu Y, Ding Y, Zhang Z, Gao Y. Exploring Endothelial Cell Dysfunction's Impact on the Brain-Retina Microenvironment Connection: Molecular Mechanisms and Implications. Mol Neurobiol 2025:10.1007/s12035-025-04714-x. [PMID: 39904964 DOI: 10.1007/s12035-025-04714-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 01/19/2025] [Indexed: 02/06/2025]
Abstract
The intricate linking between the health of blood vessels and the functioning of neurons has attracted growing attention in the context of disorders that affect the neurological environment. Endothelial cells, forming the blood-brain barrier and blood-retinal barrier, play a fundamental role in maintaining the integrity of the brain-retina microenvironment connection. This review explores the molecular foundations of endothelial cell dysfunction and its implications for the brain-retina interaction. A comprehensive analysis of the complex factors contributing to endothelial dysfunction is presented, including oxidative stress, inflammation, reduced nitric oxide signaling, and disrupted vascular autoregulation. The significance of endothelial dysfunction extends to neurovascular coupling, synaptic plasticity, and trophic support. To our knowledge, there is currently no existing literature review addressing endothelial microvascular dysfunction and its interplay with the brain-retina microenvironment. The review also explains bidirectional communication between the brain and retina, highlighting how compromised endothelial function can disrupt this vital crosstalk and inhibit normal physiological processes. As neurodegenerative diseases frequently exhibit vascular involvement, a deeper comprehension of the interaction between endothelial cells and neural tissue holds promise for innovative therapeutic strategies. By targeting endothelial dysfunction, we may enhance our ability to preserve the intricate dynamics of the brain-retina microenvironment connection and ameliorate the progression of neurological disorders.
Collapse
Affiliation(s)
- Wahid Shah
- Shanxi Eye Hospital Affiliated to Shanxi Medical University, Taiyuan, 030001, China
- Shanxi Eye Hospital Affiliated to Shanxi Medical University, Taiyuan, 030002, China
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, 030001, China
| | - Yuxing Gong
- Shanxi Eye Hospital Affiliated to Shanxi Medical University, Taiyuan, 030001, China
| | - Xin Qiao
- Shanxi Eye Hospital Affiliated to Shanxi Medical University, Taiyuan, 030002, China
| | - Yaling Lu
- Shanxi Eye Hospital Affiliated to Shanxi Medical University, Taiyuan, 030002, China
| | - Yufei Ding
- Shanxi Eye Hospital Affiliated to Shanxi Medical University, Taiyuan, 030002, China
| | - Ziting Zhang
- Shanxi Eye Hospital Affiliated to Shanxi Medical University, Taiyuan, 030002, China
| | - Yuan Gao
- Shanxi Eye Hospital Affiliated to Shanxi Medical University, Taiyuan, 030002, China.
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, 030001, China.
| |
Collapse
|
6
|
Husain A, Winebrake JP, Goldberg NR, Mahrous MA, Kovacs KD. Delayed-Onset White-Dot Syndrome in the Setting of Traumatic Choroidal Rupture. JOURNAL OF VITREORETINAL DISEASES 2025:24741264251315157. [PMID: 39897624 PMCID: PMC11783407 DOI: 10.1177/24741264251315157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Purpose To describe a patient with a traumatic choroidal rupture and a submacular hemorrhage, the course of which was complicated by delayed-onset posterior uveitis resembling a white-dot syndrome. Methods A single case was evaluated. Results A 34-year-old man presented after being struck in the left eye with a tennis ball. The visual acuity (VA) was 20/30 with otherwise normal ophthalmic vitals. An examination showed traumatic iritis and choroidal rupture with a submacular hemorrhage without subfoveal involvement. Despite treatment of anterior segment inflammation and a worsening hemorrhage with topical agents and intravitreal aflibercept, the VA decreased to 20/600. A repeat examination with optical coherence tomography showed new optic disc edema, placoid outer retinal lesions adjacent to the choroidal rupture, and corresponding ellipsoid zone atrophy. A broad workup was unremarkable, and the patient completed a long taper of high-dose oral prednisone without recurrence. Conclusions Traumatic exposure of the immunologically privileged subretinal space to high-flow choroidal circulation likely triggered a pathway involving self-autoantigenicity and a uveitic response.
Collapse
Affiliation(s)
- Alina Husain
- Department of Ophthalmology, and MD Program, Weill Cornell Medicine, New York, NY, USA
| | | | - Naomi R. Goldberg
- Department of Ophthalmology, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Great Neck, NY, USA
| | | | - Kyle D. Kovacs
- Department of Ophthalmology, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
7
|
Lavekar SS, Hughes JM, Gomes C, Huang KC, Harkin J, Canfield SG, Meyer JS. Exploring dysfunctional barrier phenotypes associated with glaucoma using a human pluripotent stem cell-based model of the neurovascular unit. Fluids Barriers CNS 2024; 21:90. [PMID: 39543684 PMCID: PMC11566410 DOI: 10.1186/s12987-024-00593-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 11/03/2024] [Indexed: 11/17/2024] Open
Abstract
Glaucoma is a neurodegenerative disease that results in the degeneration of retinal ganglion cells (RGCs) and subsequent loss of vision. While RGCs are the primary cell type affected in glaucoma, neighboring cell types selectively modulate RGCs to maintain overall homeostasis. Among these neighboring cell types, astrocytes, microvascular endothelial cells (MVECs), and pericytes coordinate with neurons to form the neurovascular unit that provides a physical barrier to limit the passage of toxic materials from the blood into neural tissue. Previous studies have demonstrated that these barrier properties may be compromised in the progression of glaucoma, yet mechanisms by which this happens have remained incompletely understood. Thus, the goals of this study were to adapt a human pluripotent stem cell (hPSC)-based model of the neurovascular unit to the study of barrier integrity relevant to glaucoma. To achieve this, hPSCs were differentiated into the cell types that contribute to this barrier, including RGCs, astrocytes, and MVECs, then assembled into an established Transwell®-insert model. The ability of these cell types to contribute to an in vitro barrier model was tested for their ability to recapitulate characteristic barrier properties. Results revealed that barrier properties of MVECs were enhanced when cultured in the presence of RGCs and astrocytes compared to MVECs cultured alone. Conversely, the versatility of this system to model aspects of barrier dysfunction relevant to glaucoma was tested using an hPSC line with a glaucoma-specific Optineurin (E50K) mutation as well as a paired isogenic control, where MVECs then exhibited reduced barrier integrity. To identify factors that could result in barrier dysfunction, results revealed an increased expression of TGFβ2 in glaucoma-associated OPTN(E50K) astrocytes, indicating a potential role for TGFβ2 in disease manifestation. To test this hypothesis, we explored the ability to modulate exogenous TGFβ2 in both isogenic control and OPTN(E50K) experimental conditions. Collectively, the results of this study indicated that the repurposing of this in vitro barrier model for glaucoma reliably mimicked some aspects of barrier dysfunction, and may serve as a platform for drug discovery, as well as a powerful in vitro model to test the consequences of barrier dysfunction upon RGCs in glaucoma.
Collapse
Affiliation(s)
- Sailee S Lavekar
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Jason M Hughes
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Terre Haute, IN, 47809, USA
| | - Cátia Gomes
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Kang-Chieh Huang
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Jade Harkin
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Scott G Canfield
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Terre Haute, IN, 47809, USA.
| | - Jason S Meyer
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
- Department of Ophthalmology, Glick Eye Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| |
Collapse
|
8
|
Wang S, Tong S, Jin X, Li N, Dang P, Sui Y, Liu Y, Wang D. Single-cell RNA sequencing analysis of the retina under acute high intraocular pressure. Neural Regen Res 2024; 19:2522-2531. [PMID: 38526288 PMCID: PMC11090430 DOI: 10.4103/1673-5374.389363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 07/27/2023] [Accepted: 09/13/2023] [Indexed: 03/26/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202419110-00032/figure1/v/2024-03-08T184507Z/r/image-tiff High intraocular pressure causes retinal ganglion cell injury in primary and secondary glaucoma diseases, yet the molecular landscape characteristics of retinal cells under high intraocular pressure remain unknown. Rat models of acute hypertension ocular pressure were established by injection of cross-linked hyaluronic acid hydrogel (Healaflow®). Single-cell RNA sequencing was then used to describe the cellular composition and molecular profile of the retina following high intraocular pressure. Our results identified a total of 12 cell types, namely retinal pigment epithelial cells, rod-photoreceptor cells, bipolar cells, Müller cells, microglia, cone-photoreceptor cells, retinal ganglion cells, endothelial cells, retinal progenitor cells, oligodendrocytes, pericytes, and fibroblasts. The single-cell RNA sequencing analysis of the retina under acute high intraocular pressure revealed obvious changes in the proportions of various retinal cells, with ganglion cells decreased by 23%. Hematoxylin and eosin staining and TUNEL staining confirmed the damage to retinal ganglion cells under high intraocular pressure. We extracted data from retinal ganglion cells and analyzed the retinal ganglion cell cluster with the most distinct expression. We found upregulation of the B3gat2 gene, which is associated with neuronal migration and adhesion, and downregulation of the Tsc22d gene, which participates in inhibition of inflammation. This study is the first to reveal molecular changes and intercellular interactions in the retina under high intraocular pressure. These data contribute to understanding of the molecular mechanism of retinal injury induced by high intraocular pressure and will benefit the development of novel therapies.
Collapse
Affiliation(s)
- Shaojun Wang
- Division of Ophthalmology, The Third Medical Center of PLA General Hospital, Beijing, China
| | - Siti Tong
- Division of Ophthalmology, The Third Medical Center of PLA General Hospital, Beijing, China
| | - Xin Jin
- Division of Ophthalmology, The Third Medical Center of PLA General Hospital, Beijing, China
| | - Na Li
- Division of Ophthalmology, The Third Medical Center of PLA General Hospital, Beijing, China
| | - Pingxiu Dang
- Division of Ophthalmology, The Third Medical Center of PLA General Hospital, Beijing, China
| | - Yang Sui
- Division of Ophthalmology, The Third Medical Center of PLA General Hospital, Beijing, China
| | - Ying Liu
- Department of Ophthalmology, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
| | - Dajiang Wang
- Division of Ophthalmology, The Third Medical Center of PLA General Hospital, Beijing, China
| |
Collapse
|
9
|
Ullah Z, Tao Y, Mehmood A, Huang J. The Role of Gut Microbiota in the Pathogenesis of Glaucoma: Evidence from Bibliometric Analysis and Comprehensive Review. Bioengineering (Basel) 2024; 11:1063. [PMID: 39593723 PMCID: PMC11591249 DOI: 10.3390/bioengineering11111063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/08/2024] [Accepted: 10/19/2024] [Indexed: 11/28/2024] Open
Abstract
The relationship between gut microbiota and glaucoma has garnered significant interest, with emerging evidence suggesting that gut dysbiosis, inflammation, and immune mechanisms may contribute to glaucoma pathogenesis. Understanding these interactions through the gut-retina axis offers new insights into disease progression and potential therapeutic options. This study combines bibliometric analysis and literature review to evaluate research trends and key research areas related to gut microbiota's role in glaucoma. Our data were collected from the Web of Science Core Collection (WoSCC) and included the English original articles and reviews published between 1 January 2008, and 6 August 2024. Visual and statistical analyses were conducted using VOSviewer and CiteSpace. The analyses comprised 810 citations from leading journals, representing contributions from 23 countries/regions, 111 institutions, 40 journals, and 321 authors. Among the countries and regions involved, the USA and China were the leading contributors, publishing the most articles and being major research hubs. The Experimental Eye Research and Investigative Ophthalmology & Visual Science were the top journals in citation and co-citations that produced high-quality publications. The top 10 highly cited articles were published in high-ranking, top-quartile journals. The frequently occurring keywords were "glaucoma", "microbiota", "gut microbiota", "inflammation", "gut-retina axis", and "probiotics". Our study highlights the growing interest in the association between gut microbiota and glaucoma. It summarizes the possible ways gut microbiota dysbiosis, systemic and neuroinflammation, and autoimmune mechanisms contribute to glaucomatous pathogenesis. Future research should focus on mechanistic studies to elucidate the pathways linking gut microbiota to glaucoma development and progression.
Collapse
Affiliation(s)
- Zakir Ullah
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha 410017, China; (Z.U.); (Y.T.)
- Taiwan International Graduate Program in Interdisciplinary Neuroscience, National Cheng Kung University and Academia Sinica, Taipei 115014, Taiwan
| | - Yuanyuan Tao
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha 410017, China; (Z.U.); (Y.T.)
| | - Amina Mehmood
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan 701401, Taiwan;
| | - Jufang Huang
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha 410017, China; (Z.U.); (Y.T.)
| |
Collapse
|
10
|
Zhang C, Simón M, Lim H, Tolman NG, Horbal L, Juarez FA, Bhandari A, Montgomery C, John SWM. IOP-induced blood-retinal barrier compromise contributes to RGC death in glaucoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.15.618539. [PMID: 39463998 PMCID: PMC11507889 DOI: 10.1101/2024.10.15.618539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
The integrity of the blood-retinal barrier (BRB) has been largely unexplored in glaucoma. We reveal that elevated intraocular pressure (IOP) partially compromises the BRB in two human-relevant inherited mouse models of glaucoma (DBA/2J and Lmx1bV265D). Experimentally increasing IOP in mouse eyes further confirms this. Notably, the compromise induces subtle leakage, happening without bleeding or detected endothelial cell junction disruption, and it precedes neurodegeneration. Leakage occurs from peripheral veins in the retinal ganglion cell layer with a concomitant loss of the transcytosis inhibitor MFSD2A. Importantly, stabilizing β-catenin in retinal endothelial cells prevents both vascular leakage and neurodegeneration in the DBA/2J model. The occurrence of leakage in all 3 high IOP models indicates that BRB compromise may be a common, yet overlooked, mechanism in glaucoma. These findings suggest that IOP-induced BRB compromise plays a critical role in glaucoma, offering a new therapeutic target.
Collapse
|
11
|
Wu X, Zhu H, Liu J, Ouyang S, Lyu Z, Jin Y, Chen X, Meng Q. Jagged1-Notch1 Signaling Pathway Induces M1 Microglia to Disrupt the Barrier Function of Retinal Microvascular Endothelial Cells. Curr Eye Res 2024; 49:1098-1106. [PMID: 38783634 DOI: 10.1080/02713683.2024.2357601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 04/28/2024] [Accepted: 05/14/2024] [Indexed: 05/25/2024]
Abstract
PURPOSE Microglia-related inflammation is closely linked to the pathogenesis of retinal diseases. The primary objective of this research was to investigate the impact and mechanism of M1 phenotype microglia on the barrier function of retina microvascular endothelial cells. METHODS Quantitative polymerase chain reactions and western blot techniques were utilized to analysis the mRNA and protein expressions of M1 and M2 markers of human microglial clone 3 cell line (HMC3), as well as the levels of Notch ligands and receptors under the intervention of lipopolysaccharide (LPS) or interleukin (IL)-4. ELISA was utilized to detect the pro-inflammatory and anti-inflammatory cytokines from HMC3 cells. The cellular tight junction and apoptosis of human retinal microvascular endothelial cells (HRMECs) were assessed by western blot and fluorescein isothiocyanate-dextran permeability assay. The inhibitors of Notch1 and RNA interference (RNAi) targeting Jagged1 were used to assess their contribution to the barrier function of vascular endothelial cells. RESULTS Inducible nitric oxide synthase (iNOS) and IL-1β were considerably elevated in LPS-treated HMC3, while CD206 and Arg-1 markedly elevated under IL-4 stimulation. The conditioned medium derived from LPS-treated HMC3 cells promoted permeability, diminished the expression of zonula occludens-1 and Occludin, and elevated the expression of Cleaved caspase-3 in HRMECs. RNAi targeting Jagged1 or Notch1 inhibitor could block M1 HMC3 polarization and maintain barrier function of HRMECs. CONCLUSION Our findings suggest that Jagged1-Notch1 signaling pathway induces M1 microglial cells to disrupt the barrier function of HRMECs, which may lead to retinal diseases.
Collapse
Affiliation(s)
- Xiyu Wu
- School of Medicine, South China University of Technology, Guangzhou, China
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Haoxian Zhu
- School of Medicine, South China University of Technology, Guangzhou, China
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Junbin Liu
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Shuyi Ouyang
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Zheng Lyu
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yeanqi Jin
- School of Medicine, South China University of Technology, Guangzhou, China
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Xinyu Chen
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Qianli Meng
- School of Medicine, South China University of Technology, Guangzhou, China
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| |
Collapse
|
12
|
Xu X, Xu S, Gao Y, He S, He J, Chen X, Guo J, Zhang X. Remote ischemic conditioning slows blood-retinal barrier damage in type 1 diabetic rats. Brain Res 2024; 1846:149253. [PMID: 39332618 DOI: 10.1016/j.brainres.2024.149253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 09/03/2024] [Accepted: 09/24/2024] [Indexed: 09/29/2024]
Abstract
Diabetic retinopathy (DR) is one of the major complications of diabetes and can cause severe visual impairment. Blood-retina barrier (BRB) destruction resulted from chronic hyperglycemia underlines its major pathological process. However, current treatments have limited efficacy and may even cause serious complications. Remote ischemic conditioning (RIC), through repeated transient mechanical occlusion of limb blood vessels, has been confirmed to promote blood-brain barrier integrity after stroke, but its role in BRB disruption has not been elucidated. This study aimed to investigate the protective effects of RIC on the BRB in diabetic rats and its potential mechanisms. 48 Sprague-Dawley rats were randomly assigned to the Sham group, Sham + RIC group, diabetes mellitus (DM) group and DM+RIC group. The diabetic model was successfully induced by intraperitoneal injection of streptozotocin. RIC treatment was administered daily and lasted for 9 weeks. In functional analysis, RIC improved the retinal function based on electroretinogram data and reduced the leakage of BRB in diabetic rats. In proteomic analysis, tight junction pathway was enriched after RIC treatment, in which Patj gene was significantly increased. We also found that RIC increased mRNA levels of Patj, claudin-1 and zonula occludens (ZO)-1, protein expression of claudin-1 when compared with diabetic models. In conclusion, RIC slowed BRB damage in diabetic rats, which may be related to the preservation of tight junction proteins. RIC may be a promising protective strategy for the treatment of DR.
Collapse
Affiliation(s)
- Xiaohan Xu
- Department of Ophthalmology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Shuaili Xu
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Yuan Gao
- Department of Ophthalmology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Shan He
- Department of Ophthalmology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Jiachen He
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Xi Chen
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Jiaqi Guo
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Xuxiang Zhang
- Department of Ophthalmology, Xuanwu Hospital of Capital Medical University, Beijing, China.
| |
Collapse
|
13
|
Kim SY, Cheon J. Senescence-associated microvascular endothelial dysfunction: A focus on the blood-brain and blood-retinal barriers. Ageing Res Rev 2024; 100:102446. [PMID: 39111407 DOI: 10.1016/j.arr.2024.102446] [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: 03/08/2024] [Revised: 07/05/2024] [Accepted: 08/02/2024] [Indexed: 08/17/2024]
Abstract
The blood-brain barrier (BBB) and blood-retinal barrier (BRB) constitute critical physiochemical interfaces, precisely orchestrating the bidirectional communication between the brain/retina and blood. Increased permeability or leakage of these barriers has been demonstrably linked to age-related vascular and parenchymal damage. While it has been suggested that the gradual aging process may coincide with disruptions in these barriers, this phenomenon is significantly exacerbated in individuals with age-related neurodegenerative disorders (ARND). This review focuses on the microvascular endothelium, a key constituent of BBB and BRB, highlighting the impact of endothelial senescence on barrier dysfunction and exploring recent discoveries regarding core pathways implicated in its breakdown. Subsequently, we address the "vascular senescence hypothesis" for ARND, with a particular emphasis on Alzheimer's disease and age-related macular degeneration, centered on endothelial senescence. Finally, we discuss potential senotherapeutic strategies targeting barrier dysfunction.
Collapse
Affiliation(s)
- Sung Young Kim
- Department of Biochemistry, Konkuk University School of Medicine, Republic of Korea; Research Institute of Medical Science, Konkuk University, Republic of Korea; IBST, Konkuk University, Republic of Korea.
| | - Jaejoung Cheon
- Department of Biochemistry, Konkuk University School of Medicine, Republic of Korea
| |
Collapse
|
14
|
Zhou Y, Zhao C, Shi Z, Heger Z, Jing H, Shi Z, Dou Y, Wang S, Qiu Z, Li N. A Glucose-Responsive Hydrogel Inhibits Primary and Secondary BRB Injury for Retinal Microenvironment Remodeling in Diabetic Retinopathy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402368. [PMID: 39031576 PMCID: PMC11348052 DOI: 10.1002/advs.202402368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 06/06/2024] [Indexed: 07/22/2024]
Abstract
Current diabetic retinopathy (DR) treatment involves blood glucose regulation combined with laser photocoagulation or intravitreal injection of vascular endothelial growth factor (VEGF) antibodies. However, due to the complex pathogenesis and cross-interference of multiple biochemical pathways, these interventions cannot block disease progression. Recognizing the critical role of the retinal microenvironment (RME) in DR, it is hypothesized that reshaping the RME by simultaneously inhibiting primary and secondary blood-retinal barrier (BRB) injury can attenuate DR. For this, a glucose-responsive hydrogel named Cu-PEI/siMyD88@GEMA-Con A (CSGC) is developed that effectively delivers Cu-PEI/siMyD88 nanoparticles (NPs) to the retinal pigment epithelium (RPE). The Cu-PEI NPs act as antioxidant enzymes, scavenging ROS and inhibiting RPE pyroptosis, ultimately blocking primary BRB injury by reducing microglial activation and Th1 differentiation. Simultaneously, MyD88 expression silence in combination with the Cu-PEI NPs decreases IL-18 production, synergistically reduces VEGF levels, and enhances tight junction proteins expression, thus blocking secondary BRB injury. In summary, via remodeling the RME, the CSGC hydrogel has the potential to disrupt the detrimental cycle of cross-interference between primary and secondary BRB injury, providing a promising therapeutic strategy for DR.
Collapse
Affiliation(s)
- Yue Zhou
- Tianjin Key Laboratory of Drug Delivery & High‐EfficiencySchool of Pharmaceutical Science and TechnologyTianjin UniversityTianjin300072P. R. China
- Department of PharmacyTianjin Union Medical CenterNankai UniversityTianjin300122P. R. China
| | - Chan Zhao
- Department of OphthalmologyPeking Union Medical College HospitalChinese Academy of Medical SciencesBeijing100730P. R. China
- Key Laboratory of Ocular Fundus DiseasesChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100730P. R. China
| | - Zhiyuan Shi
- Tianjin Key Laboratory of Drug Delivery & High‐EfficiencySchool of Pharmaceutical Science and TechnologyTianjin UniversityTianjin300072P. R. China
| | - Zbynek Heger
- Department of Chemistry and BiochemistryMendel University in BrnoBrnoCZ‐61300Czech Republic
| | - HuaQing Jing
- Tianjin Key Laboratory of Drug Delivery & High‐EfficiencySchool of Pharmaceutical Science and TechnologyTianjin UniversityTianjin300072P. R. China
| | - Zhengming Shi
- Department of OphthalmologyPeking Union Medical College HospitalChinese Academy of Medical SciencesBeijing100730P. R. China
- Key Laboratory of Ocular Fundus DiseasesChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100730P. R. China
| | - Yunsheng Dou
- Tianjin Key Laboratory of Drug Delivery & High‐EfficiencySchool of Pharmaceutical Science and TechnologyTianjin UniversityTianjin300072P. R. China
| | - Siyu Wang
- Tianjin Key Laboratory of Drug Delivery & High‐EfficiencySchool of Pharmaceutical Science and TechnologyTianjin UniversityTianjin300072P. R. China
| | - Zitong Qiu
- Tianjin Key Laboratory of Drug Delivery & High‐EfficiencySchool of Pharmaceutical Science and TechnologyTianjin UniversityTianjin300072P. R. China
| | - Nan Li
- Tianjin Key Laboratory of Drug Delivery & High‐EfficiencySchool of Pharmaceutical Science and TechnologyTianjin UniversityTianjin300072P. R. China
| |
Collapse
|
15
|
Wang XF, Xiang XH, Wei J, Zhang PB, Xu Q, Liu MH, Qu LQ, Wang XX, Yu L, Wu AG, Qing DL, Wu JM, Law BYK, Yu CL, Yong-Tang. Raddeanin A Protects the BRB Through Inhibiting Inflammation and Apoptosis in the Retina of Alzheimer's Disease. Neurochem Res 2024; 49:2197-2214. [PMID: 38834846 DOI: 10.1007/s11064-024-04145-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 02/28/2024] [Accepted: 05/02/2024] [Indexed: 06/06/2024]
Abstract
Neuroinflammation and endothelial cell apoptosis are prominent features of blood-brain barrier (BBB) disruption, which have been described in Alzheimer's disease (AD) and can predict cognitive decline. Recent reports revealed vascular β-amyloid (Aβ) deposits, Muller cell degeneration and microglial dysfunction in the retina of AD patients. However, there has been no in-depth research on the roles of inflammation, retinal endothelial cell apoptosis, and blood-retinal barrier (BRB) damage in AD retinopathy. We found that Raddeanin A (RDA) could improve pathological and cognitive deficits in a mouse model of Alzheimer's disease by targeting β-amyloidosis, However, the effects of RDA on AD retinal function require further study. To clarify whether RDA inhibits inflammation and apoptosis and thus improves BRB function in AD-related retinopathy. In vitro we used Aβ-treated HRECs and MIO-M1 cells, and in vivo we used 3×Tg-AD mice to investigate the effect of RDA on BRB in AD-related retinopathy. We found that RDA could improve BRB function in AD-related retinopathy by inhibiting NLRP3-mediated inflammation and suppressing Wnt/β-catenin pathway-mediated apoptosis, which is expected to improve the pathological changes in AD-related retinopathy and the quality of life of AD patients.
Collapse
Affiliation(s)
- Xiao-Fang Wang
- Laboratory Animal Centre, Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
- School of Basic Medical Sciences of Southwest Medical University, Luzhou, China
| | - Xiao-Hong Xiang
- Department of Ophthalmology in The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Jing Wei
- Eye School and Key Laboratory of Sichuan Province Ophthalmopathy Prevention & Cure and Visual Function Protection of Chengdu University of TCM, Chengdu, China
| | - Peng-Bo Zhang
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, 15651, China
| | - Qin Xu
- Department of Ophthalmology in The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Meng-Han Liu
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine and Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Li-Qun Qu
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine and Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Xing-Xia Wang
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine and Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Lu Yu
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica of Southwest Medical University, Luzhou, 646000, China
| | - An-Guo Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica of Southwest Medical University, Luzhou, 646000, China
| | - Da-Lian Qing
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica of Southwest Medical University, Luzhou, 646000, China
| | - Jian-Ming Wu
- School of Basic Medical Sciences of Southwest Medical University, Luzhou, China
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica of Southwest Medical University, Luzhou, 646000, China
| | - Betty Yuen-Kwan Law
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine and Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China.
| | - Chong-Lin Yu
- School of Basic Medical Sciences of Southwest Medical University, Luzhou, China.
| | - Yong-Tang
- Laboratory Animal Centre, Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China.
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine and Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China.
- School of Basic Medical Sciences of Southwest Medical University, Luzhou, China.
| |
Collapse
|
16
|
Sharma M, Pal P, Gupta SK. Microglial mediators in autoimmune Uveitis: Bridging neuroprotection and neurotoxicity. Int Immunopharmacol 2024; 136:112309. [PMID: 38810304 DOI: 10.1016/j.intimp.2024.112309] [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: 01/24/2024] [Revised: 04/29/2024] [Accepted: 05/16/2024] [Indexed: 05/31/2024]
Abstract
Autoimmune uveitis, a severe inflammatory condition of the eye, poses significant challenges due to its complex pathophysiology and the critical balance between protective and detrimental immune responses. Central to this balance are microglia, the resident immune cells of the central nervous system, whose roles in autoimmune uveitis are multifaceted and dynamic. This review article delves into the dual nature of microglial functions, oscillating between neuroprotective and neurotoxic outcomes in the context of autoimmune uveitis. Initially, we explore the fundamental aspects of microglia, including their activation states and basic functions, setting the stage for a deeper understanding of their involvement in autoimmune uveitis. The review then navigates through the intricate mechanisms by which microglia contribute to disease onset and progression, highlighting both their protective actions in immune regulation and tissue repair, and their shift towards a pro-inflammatory, neurotoxic profile. Special emphasis is placed on the detailed pathways and cellular interactions underpinning these dual roles. Additionally, the review examines the potential of microglial markers as diagnostic and prognostic indicators, offering insights into their clinical relevance. The article culminates in discussing future research directions, and the ongoing challenges in translating these findings into effective clinical applications. By providing a comprehensive overview of microglial mechanisms in autoimmune uveitis, this review underscores the critical balance of microglial activities and its implications for disease management and therapy development.
Collapse
Affiliation(s)
- Monika Sharma
- Department of Pharmacology, Faculty of Pharmacy, Swami Vivekanand Subharti University, Meerut, Uttar Pradesh, India
| | - Pankaj Pal
- Department of Pharmacy, Banasthali Vidyapith, Rajasthan, India.
| | - Sukesh Kumar Gupta
- KIET School of Pharmacy, KIET Group of Institutions, Ghaziabad, Uttar Pradesh, India; Department of Ophthalmology, Visual and Anatomical Sciences (OVAS), School of Medicine, Wayne State University, USA.
| |
Collapse
|
17
|
Nguyen Y, Rudd Zhong Manis J, Ronczkowski NM, Bui T, Oxenrider A, Jadeja RN, Thounaojam MC. Unveiling the gut-eye axis: how microbial metabolites influence ocular health and disease. Front Med (Lausanne) 2024; 11:1377186. [PMID: 38799150 PMCID: PMC11122920 DOI: 10.3389/fmed.2024.1377186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 04/19/2024] [Indexed: 05/29/2024] Open
Abstract
The intricate interplay between the gut microbiota and ocular health has surpassed conventional medical beliefs, fundamentally reshaping our understanding of organ interconnectivity. This review investigates into the intricate relationship between gut microbiota-derived metabolites and their consequential impact on ocular health and disease pathogenesis. By examining the role of specific metabolites, such as short-chain fatty acids (SCFAs) like butyrate and bile acids (BAs), herein we elucidate their significant contributions to ocular pathologies, thought-provoking the traditional belief of organ sterility, particularly in the field of ophthalmology. Highlighting the dynamic nature of the gut microbiota and its profound influence on ocular health, this review underlines the necessity of comprehending the complex workings of the gut-eye axis, an emerging field of science ready for further exploration and scrutiny. While acknowledging the therapeutic promise in manipulating the gut microbiome and its metabolites, the available literature advocates for a targeted, precise approach. Instead of broad interventions, it emphasizes the potential of exploiting specific microbiome-related metabolites as a focused strategy. This targeted approach compared to a precision tool rather than a broad-spectrum solution, aims to explore the therapeutic applications of microbiome-related metabolites in the context of various retinal diseases. By proposing a nuanced strategy targeted at specific microbial metabolites, this review suggests that addressing specific deficiencies or imbalances through microbiome-related metabolites might yield expedited and pronounced outcomes in systemic health, extending to the eye. This focused strategy holds the potential in bypassing the irregularity associated with manipulating microbes themselves, paving a more efficient pathway toward desired outcomes in optimizing gut health and its implications for retinal diseases.
Collapse
Affiliation(s)
- Yvonne Nguyen
- Mercer University School of Medicine, Macon, GA, United States
| | | | | | - Tommy Bui
- Departments of Cellular Biology and Anatomy, Augusta University, Augusta, GA, United States
| | - Allston Oxenrider
- Departments of Cellular Biology and Anatomy, Augusta University, Augusta, GA, United States
| | - Ravirajsinh N. Jadeja
- Biochemistry and Molecular Biology, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Menaka C. Thounaojam
- Departments of Cellular Biology and Anatomy, Augusta University, Augusta, GA, United States
| |
Collapse
|
18
|
Buthelezi LM, Munsamy AJ, Mashige KP. Inflammatory mechanisms contributing to retinal alterations in HIV infection and long-term ART. South Afr J HIV Med 2024; 25:1548. [PMID: 38628910 PMCID: PMC11019112 DOI: 10.4102/sajhivmed.v25i1.1548] [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: 10/30/2023] [Accepted: 01/24/2024] [Indexed: 04/19/2024] Open
Abstract
People living with HIV (PLWH) may face an increased risk of eye complications associated with ageing, chronic inflammation, and the toxicity arising from long-term antiretroviral therapy (ART). This review aims to understand how inflammatory pathways contribute to retinal alterations observed in PLWH on long-term ART. This review was conducted using four electronic database searches, namely Scopus, Hinari, Google Scholar, and PubMed; from 1996 (when ART became available) until January 2022, without language restriction. Sources from clinical trials, meta-analyses, randomised controlled trials, and systematic reviews were used. Dysregulated para-inflammation (chronic inflammation) damages the blood-retina barrier, resulting in the altered retinal immune privilege and leading to the development of retinal and blood vessel changes. There is an interplay between the effects of the disease versus ART. ART causes mitochondrial toxicity, which affects the retinal ganglion cells and retinal pigment epithelium (RPE) due to oxidative stress. Infection by HIV also affects retinal microglia, which contributes to RPE damage. Both of these mechanisms affect the blood vessels. Assessing the integrity of the inner and outer blood-retina barrier is a pivotal point in pinpointing the pathogenesis of inner retinal alterations. Optical coherence tomography is a valuable tool to assess these changes. There is a paucity of research to understand how these structural changes may affect visual function, such as contrast sensitivity and colour vision.
Collapse
Affiliation(s)
- Lungile M Buthelezi
- Department of Optometry, School of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Alvin J Munsamy
- Department of Optometry, School of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Khathutshelo P Mashige
- Department of Optometry, School of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
19
|
Tomkins-Netzer O, Niederer R, Greenwood J, Fabian ID, Serlin Y, Friedman A, Lightman S. Mechanisms of blood-retinal barrier disruption related to intraocular inflammation and malignancy. Prog Retin Eye Res 2024; 99:101245. [PMID: 38242492 DOI: 10.1016/j.preteyeres.2024.101245] [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: 09/29/2023] [Revised: 01/16/2024] [Accepted: 01/16/2024] [Indexed: 01/21/2024]
Abstract
Blood-retinal barrier (BRB) disruption is a common accompaniment of intermediate, posterior and panuveitis causing leakage into the retina and macular oedema resulting in vision loss. It is much less common in anterior uveitis or in patients with intraocular lymphoma who may have marked signs of intraocular inflammation. New drugs used for chemotherapy (cytarabine, immune checkpoint inhibitors, BRAF inhibitors, EGFR inhibitors, bispecific anti-EGFR inhibitors, MET receptor inhibitors and Bruton tyrosine kinase inhibitors) can also cause different types of uveitis and BRB disruption. As malignant disease itself can cause uveitis, particularly from breast, lung and gastrointestinal tract cancers, it can be clinically difficult to sort out the cause of BRB disruption. Immunosuppression due to malignant disease and/or chemotherapy can lead to infection which can also cause BRB disruption and intraocular infection. In this paper we address the pathophysiology of BRB disruption related to intraocular inflammation and malignancy, methods for estimating the extent and effect of the disruption and examine why some types of intraocular inflammation and malignancy cause BRB disruption and others do not. Understanding this may help sort and manage these patients, as well as devise future therapeutic approaches.
Collapse
Affiliation(s)
- Oren Tomkins-Netzer
- Department of Ophthalmology, Lady Davis Carmel Medical Centre, Haifa, Israel; Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel.
| | - Rachael Niederer
- Department of Ophthalmology, Te Whatu Ora, Auckland, New Zealand; Department of Ophthalmology, University of Auckland, Auckland, New Zealand
| | - John Greenwood
- Institute of Ophthalmology, University College London, London, UK
| | - Ido Didi Fabian
- The Goldschleger Eye Institute, Sheba Medical Centre, Tel Hashomer, Tel Aviv University, Tel Aviv, Israel
| | - Yonatan Serlin
- Department of Medical Neuroscience and the Brain Repair Centre, Dalhousie University, Faculty of Medicine, Halifax, NS, Canada
| | - Alon Friedman
- Department of Medical Neuroscience and the Brain Repair Centre, Dalhousie University, Faculty of Medicine, Halifax, NS, Canada; Departments of Physiology and Cell Biology, Brain and Cognitive Sciences, Zlotowski Centre for Neuroscience, Ben- Gurion University of the Negev, Beer-Sheva, Israel
| | - Sue Lightman
- Institute of Ophthalmology, University College London, London, UK
| |
Collapse
|
20
|
Adeeb S, Arabi TZ, Shah H, Alsalameh S, Abu-Shaar M, El-Sibai AM, Alkattan K, Yaqinuddin A. Unveiling the Web: Exploring the Multifaceted Role of Neutrophil Extracellular Traps in Ocular Health and Disease. J Clin Med 2024; 13:512. [PMID: 38256646 PMCID: PMC10816449 DOI: 10.3390/jcm13020512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 01/02/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Neutrophil extracellular traps (NETs) play an essential role in antimicrobial defense. However, NETs have also been shown to promote and mediate a wide spectrum of diseases, including cancer, diabetes mellitus, cardiovascular diseases, and ocular diseases. Data regarding NETs in ocular diseases remain limited. In physiological conditions, NETs protect the eye from debris and cleave proinflammatory cytokines, including several interleukins. On the other hand, NETs play a role in corneal diseases, such as dry eye disease and ocular graft-versus-host disease, where they promote acinar atrophy and delayed wound healing. Additionally, NET levels positively correlate with increased severity of uveitis. NETs have also been described in the context of diabetic retinopathy. Although increased NET biomarkers are associated with an increased risk of the disease, NETs also assist in the elimination of pathological blood vessels and the regeneration of normal vessels. Targeting NET pathways for the treatment of ocular diseases has shown promising outcomes; however, more studies are still needed in this regard. In this article, we summarize the literature on the protective roles of NETs in the eye. Then, we describe their pathogenetic effects in ocular diseases, including those of the cornea, uvea, and retinal blood vessels. Finally, we describe the therapeutic implications of targeting NETs in such conditions.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Ahmed Yaqinuddin
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (S.A.); (T.Z.A.); (H.S.); (S.A.); (M.A.-S.); (A.M.E.-S.); (K.A.)
| |
Collapse
|
21
|
Yang S, Fan Z, Lu X, Liu H, Zhou Z, Qi H, Zeng J, Zheng M, Zou X, Fang S, Zhang G. Response of Human Retinal Microvascular Endothelial Cells to Influenza A (H1N1) Infection and the Underlying Molecular Mechanism. Invest Ophthalmol Vis Sci 2024; 65:38. [PMID: 38252524 PMCID: PMC10810132 DOI: 10.1167/iovs.65.1.38] [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: 10/07/2023] [Accepted: 01/03/2024] [Indexed: 01/24/2024] Open
Abstract
Purpose Whether H1N1 infection-associated ocular manifestations result from direct viral infections or systemic complications remains unclear. This study aimed to comprehensively elucidate the underlying causes and mechanism. Method TCID50 assays was performed at 24, 48, and 72 hours to verify the infection of H1N1 in human retinal microvascular endothelial cells (HRMECs). The changes in gene expression profiles of HRMECs at 24, 48, and 72 hours were characterized using RNA sequencing technology. Differentially expressed genes (DEGs) were validated using real-time quantitative polymerase chain reaction and Western blotting. CCK-8 assay and scratch assay were performed to evaluate whether there was a potential improvement of proliferation and migration in H1N1-infected cells after oseltamivir intervention. Results H1N1 can infect and replicate within HRMECs, leading to cell rounding and detachment. After H1N1 infection of HRMECs, 2562 DEGs were identified, including 1748 upregulated ones and 814 downregulated ones. These DEGs primarily involved in processes such as inflammation and immune response, cytokine-cytokine receptor interaction, signal transduction regulation, and cell adhesion. The elevated expression levels of CXCL10, CXCL11, CCL5, TLR3, C3, IFNB1, IFNG, STAT1, HLA, and TNFSF10 after H1N1 infection were reduced by oseltamivir intervention, reaching levels comparable to those in the uninfected group. The impaired cell proliferation and migration after H1N1 infection was improved by oseltamivir intervention. Conclusions This study confirmed that H1N1 can infect HRMECs, leading to the upregulation of chemokines, which may cause inflammation and destruction of the blood-retina barrier. Moreover, early oseltamivir administration may reduce retinal inflammation and hemorrhage in patients infected with H1N1.
Collapse
Affiliation(s)
- Shuo Yang
- Jinzhou Medical University, Jinzhou, Liaoning, China
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, Shenzhen, Guangdong, China
| | - Zixin Fan
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, Shenzhen, Guangdong, China
| | - Xiaofeng Lu
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, Shenzhen, Guangdong, China
| | - Hui Liu
- Shenzhen Center for Disease Control and Prevention, Shenzhen, Guangdong, China
| | - Ziying Zhou
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, Shenzhen, Guangdong, China
| | - Hui Qi
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, Shenzhen, Guangdong, China
| | - Jian Zeng
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, Shenzhen, Guangdong, China
| | - Mianying Zheng
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, Shenzhen, Guangdong, China
| | - Xuan Zou
- Shenzhen Center for Disease Control and Prevention, Shenzhen, Guangdong, China
| | - Shisong Fang
- Shenzhen Center for Disease Control and Prevention, Shenzhen, Guangdong, China
| | - Guoming Zhang
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, Shenzhen, Guangdong, China
| |
Collapse
|
22
|
Visioli G, Zeppieri M, Iannucci V, Manni P, Albanese GM, Salati C, Spadea L, Pirraglia MP. From Bedside to Diagnosis: The Role of Ocular Fundus in Systemic Infections. J Clin Med 2023; 12:7216. [PMID: 38068267 PMCID: PMC10707096 DOI: 10.3390/jcm12237216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/15/2023] [Accepted: 11/19/2023] [Indexed: 01/23/2024] Open
Abstract
In this comprehensive review, we delve into the significance of the ocular fundus examination in diagnosing and managing systemic infections at the bedside. While the utilization of advanced ophthalmological diagnostic technologies can present challenges in bedside care, especially for hospitalized patients confined to their beds or during infection outbreaks, the ocular fundus examination often emerges as an essential, and sometimes the only practical, diagnostic tool. Recent discussions have highlighted that the role of an ocular fundus examination might not always be advocated as a routine diagnostic procedure. With this context, we introduce a decision tree tailored for assessing the ocular fundus in inpatients with systemic infections. We also present an overview of systemic infections that impact the eye and elucidate key signs detectable through a bedside ocular fundus examination. Targeted primarily at non-ophthalmology clinicians, this review seeks to offer a comprehensive insight into a multifaceted approach and the enhancement of patient clinical outcomes.
Collapse
Affiliation(s)
- Giacomo Visioli
- Department of Sense Organs, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy; (G.V.)
| | - Marco Zeppieri
- Department of Ophthalmology, University Hospital of Udine, 33100 Udine, Italy
| | - Valeria Iannucci
- Department of Sense Organs, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy; (G.V.)
| | - Priscilla Manni
- Department of Sense Organs, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy; (G.V.)
| | - Giuseppe Maria Albanese
- Department of Sense Organs, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy; (G.V.)
| | - Carlo Salati
- Department of Ophthalmology, University Hospital of Udine, 33100 Udine, Italy
| | - Leopoldo Spadea
- Department of Sense Organs, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy; (G.V.)
- Eye Clinic, Policlinico Umberto I University Hospital, 00161 Rome, Italy
| | | |
Collapse
|
23
|
Zhou L, Ho BM, Chan HYE, Tong Y, Du L, He JN, Ng DSC, Tham CC, Pang CP, Chu WK. Emerging Roles of cGAS-STING Signaling in Mediating Ocular Inflammation. J Innate Immun 2023; 15:739-750. [PMID: 37778330 PMCID: PMC10616671 DOI: 10.1159/000533897] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 08/28/2023] [Indexed: 10/03/2023] Open
Abstract
Cyclic GMP-AMP (cGAMP) synthase (cGAS), a sensor of cytosolic DNA, recognizes cytoplasmic nucleic acids to activate the innate immune responses via generation of the second messenger cGAMP and subsequent activation of the stimulator of interferon genes (STINGs). The cGAS-STING signaling has multiple immunologic and physiological functions in all human vital organs. It mediates protective innate immune defense against DNA-containing pathogen infection, confers intrinsic antitumor immunity via detecting tumor-derived DNA, and gives rise to autoimmune and inflammatory diseases upon aberrant activation by cytosolic leakage of self-genomic and mitochondrial DNA. Disruptions in these functions are associated with the pathophysiology of various immunologic and neurodegenerative diseases. Recent evidence indicates important roles of the cGAS-STING signaling in mediating inflammatory responses in ocular inflammatory and inflammation-associated diseases, such as keratitis, diabetic retinopathy, age-related macular degeneration, and uveitis. In this review, we summarize the recently emerging evidence of cGAS-STING signaling in mediating ocular inflammatory responses and affecting pathogenesis of these complex eye diseases. We attempt to provide insightful perspectives on future directions of investigating cGAS-STING signaling in ocular inflammation. Understanding how cGAS-STING signaling is modulated to mediate ocular inflammatory responses would allow future development of novel therapeutic strategies to treat ocular inflammation and autoimmunity.
Collapse
Affiliation(s)
- Linbin Zhou
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR
| | - Bo Man Ho
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR
| | - Hoi Ying Emily Chan
- Medicine Programme Global Physician-Leadership Stream, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR
| | - Yan Tong
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR
| | - Lin Du
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR
| | - Jing Na He
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR
| | - Danny Siu-Chun Ng
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR
| | - Clement C. Tham
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR
| | - Chi Pui Pang
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR
| | - Wai Kit Chu
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR
- Joint Shantou International Eye Center, Shantou University and the Chinese University of Hong Kong, Shantou, China
| |
Collapse
|
24
|
Xu J, Chen L, Cen T, Zeng Y, Liu Q. HHV-6B- and HHV-7-associated choroiditis secondary to acute myelogenous leukemia: a case report. J Int Med Res 2023; 51:3000605231204479. [PMID: 37873767 PMCID: PMC10594963 DOI: 10.1177/03000605231204479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 09/13/2023] [Indexed: 10/25/2023] Open
Abstract
We report a case of human herpes virus 6 (HHV-6)- and human herpes virus 7 (HHV-7)-associated choroiditis in an immunocompromised woman. A 42-year-old Chinese woman with a history of acute myelogenous leukemia presented with blurred vision and black floaters in her right eye. Anterior segment examination findings were normal. Ophthalmoscopic examination revealed a subretinal lesion in the superonasal peripapillary region with several punctate hemorrhages. Optical coherence tomography showed a crater-like choroidal protuberance, associated with retinal pigment epithelium rupture and full-thickness retinal edema in the involved area. Indocyanine green angiography demonstrated a broad hypofluorescent lesion in the choroid. The patient was diagnosed with choroiditis. Subsequently, metagenomic next-generation sequencing revealed HHV-6B and HHV-7 DNA in the aqueous humor. Therefore, antiviral therapy was initiated. The patient experienced resolution of all symptoms and signs after treatment with intravenous foscarnet and oral acyclovir. The findings in this case indicate that HHV-6 and HHV-7 can cause ocular infection, particularly in immunocompromised patients.
Collapse
Affiliation(s)
- Jing Xu
- Department of Ophthalmology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Linjiang Chen
- Department of Ophthalmology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Tao Cen
- Department of Research and Development, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yuanping Zeng
- Department of Ophthalmology, Baiyun Branch, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Qiong Liu
- Department of Ophthalmology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
25
|
Cerna-Chavez R, Rozanska A, Poretti GL, Benvenisty N, Parulekar M, Lako M. Retinal pigment epithelium exhibits gene expression and phagocytic activity alterations when exposed to retinoblastoma chemotherapeutics. Exp Eye Res 2023; 233:109542. [PMID: 37331647 DOI: 10.1016/j.exer.2023.109542] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 01/08/2023] [Accepted: 06/15/2023] [Indexed: 06/20/2023]
Abstract
Retinoblastoma (Rb) is a rare malignant disorder affecting the developing retina of children under the age of five. Chemotherapeutic agents used for treating Rb have been associated with defects of the retinal pigment epithelium (RPE), such as hyperplasia, gliosis, and mottling. Herein, we have developed two pluripotent stem cell (PSC)-RPE models to assess the cytotoxicity of known Rb chemotherapeutics such as Melphalan, Topotecan and TW-37. Our findings demonstrate that these drugs alter the RPE by decreasing the monolayer barrier's trans-epithelial resistance and affecting the cells' phagocytic activity. Transcriptional analyses demonstrate an altered expression of genes involved in melanin and retinol processing, tight junction and apical-basal polarity pathways in both models. When applied within the clinical range, none of the drug treatments caused significant cytotoxic effects, changes to the apical-basal polarity, tight junction network or cell cycle. Together, our results demonstrate that although the most commonly used Rb chemotherapeutic drugs do not cause cytotoxicity in RPE, their application in vitro leads to compromised phagocytosis and strength of the barrier function, in addition to changes in gene expression that could alter the visual cycle in vivo. Our data demonstrate that widely used Rb chemotherapeutic drugs can have a deleterious impact on RPE cells and thus great care has to be exercised with regard to their delivery so the adjacent healthy RPE is not damaged during the course of tumor eradication.
Collapse
Affiliation(s)
| | | | | | - Nissim Benvenisty
- The Azrieli Center for Stem Cells and Genetic Research, The Hebrew University of Jerusalem, Jerusalem, Israel
| | | | | |
Collapse
|
26
|
Lepre CC, Russo M, Trotta MC, Petrillo F, D'Agostino FA, Gaudino G, D'Amico G, Campitiello MR, Crisci E, Nicoletti M, Gesualdo C, Simonelli F, D'Amico M, Hermenean A, Rossi S. Inhibition of Galectins and the P2X7 Purinergic Receptor as a Therapeutic Approach in the Neurovascular Inflammation of Diabetic Retinopathy. Int J Mol Sci 2023; 24:ijms24119721. [PMID: 37298672 DOI: 10.3390/ijms24119721] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/30/2023] [Accepted: 06/01/2023] [Indexed: 06/12/2023] Open
Abstract
Diabetic retinopathy (DR) is the most frequent microvascular retinal complication of diabetic patients, contributing to loss of vision. Recently, retinal neuroinflammation and neurodegeneration have emerged as key players in DR progression, and therefore, this review examines the neuroinflammatory molecular basis of DR. We focus on four important aspects of retinal neuroinflammation: (i) the exacerbation of endoplasmic reticulum (ER) stress; (ii) the activation of the NLRP3 inflammasome; (iii) the role of galectins; and (iv) the activation of purinergic 2X7 receptor (P2X7R). Moreover, this review proposes the selective inhibition of galectins and the P2X7R as a potential pharmacological approach to prevent the progression of DR.
Collapse
Affiliation(s)
- Caterina Claudia Lepre
- "Aurel Ardelean" Institute of Life Sciences, Vasile Goldis Western University of Arad, 310144 Arad, Romania
| | - Marina Russo
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Maria Consiglia Trotta
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Francesco Petrillo
- Ph.D. Course in Translational Medicine, Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Fabiana Anna D'Agostino
- Multidisciplinary Department of Medical, Surgical and Dental Sciences, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Gennaro Gaudino
- School of Anesthesia and Intensive Care, University of Foggia, 71122 Foggia, Italy
| | | | - Maria Rosaria Campitiello
- Department of Obstetrics and Gynecology and Physiopathology of Human Reproduction, ASL Salerno, 84124 Salerno, Italy
| | - Erminia Crisci
- Multidisciplinary Department of Medical, Surgical and Dental Sciences, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Maddalena Nicoletti
- Multidisciplinary Department of Medical, Surgical and Dental Sciences, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Carlo Gesualdo
- Multidisciplinary Department of Medical, Surgical and Dental Sciences, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Francesca Simonelli
- Multidisciplinary Department of Medical, Surgical and Dental Sciences, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Michele D'Amico
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Anca Hermenean
- "Aurel Ardelean" Institute of Life Sciences, Vasile Goldis Western University of Arad, 310144 Arad, Romania
| | - Settimio Rossi
- Multidisciplinary Department of Medical, Surgical and Dental Sciences, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| |
Collapse
|
27
|
Malan L, van Wyk R, von Känel R, Ziemssen T, Vilser W, Nilsson PM, Magnusson M, Jujic A, Mak D, Steyn F, Malan NT. The chronic stress risk phenotype mirrored in the human retina as a neurodegenerative condition. Stress 2023:1-43. [PMID: 37154816 DOI: 10.1080/10253890.2023.2210687] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/10/2023] Open
Abstract
The brain is the key organ that orchestrates the stress response which translates to the retina. The retina is an extension of the brain and retinal symptoms in subjects with neurodegenerative diseases substantiated the eye as a window to the brain. The retina is used in this study to determine whether chronic stress reflects neurodegenerative signs indicative of neurodegenerative conditions. A 3-year prospective cohort (n = 333; aged 46 ± 9 years) was stratified into stress-phenotype cases (n = 212) and controls (n = 121) by applying the Malan stress-phenotype index. Neurodegenerative risk markers included ischemia (astrocytic S100 calcium-binding protein B/S100B); 24h blood pressure, proteomics; inflammation (tumor-necrosis-factor-α/TNF-α); neuronal damage (neuron-specific-enolase); anti-apoptosis of retinal-ganglion-cells (beta-nerve-growth-factor), astrocytic activity (glial-fibrillary-acidic-protein); hematocrit (viscosity) and retinal follow-up data [vessels; stress-optic-neuropathy]. Stress-optic-neuropathy risk was calculated from two indices: a newly derived diastolic-ocular-perfusion-pressure cut-point ≥68 mmHg relating to the stress-phenotype; combined with an established cup-to-disc ratio cut-point ≥0.3. Higher stress-optic-neuropathy (39% vs. 17%) and hypertension (73% vs. 16%) prevalence was observed in the stress-phenotype cases vs. controls. Elevated diastolic-ocular-perfusion-pressure, indicating hypoperfusion, was related to arterial narrowing and trend for ischemia increases in the stress-phenotype. Ischemia in the stress-phenotype at baseline, follow-up and 3-yr changes was related to consistent inflammation (TNF-α and cytokine-interleukin-17-receptor-A), neuron-specific-enolase increases, consistent apoptosis (chitinase 3-like-1, low beta-nerve-growth-factor), glial-fibrillary-acidic-protein decreases, elevated viscosity, vein widening as risk marker of endothelial dysfunction in the blood-retinal-barrier, lower vein count, and elevated stress-optic-neuropathy. The stress-phenotype and related neurodegenerative signs of ongoing brain ischemia, apoptosis and endothelial dysfunction compromised blood-retinal-barrier permeability and optic nerve integrity. In fact, the stress-phenotype could identify persons at high risk of neurodegeneration to indicate a neurodegenerative condition.
Collapse
Affiliation(s)
- Leoné Malan
- Technology Transfer and Innovation-Support Office; Private Bag X1290, North-West University, Potchefstroom 2520, South Africa
| | - Roelof van Wyk
- Surgical Ophthalmologist; 85 Peter Mokaba Street, Potchefstroom, South Africa
| | - Roland von Känel
- Department of Consultation-Liaison Psychiatry and Psychosomatic Medicine, University Hospital Zurich; University of Zurich; Zurich Switzerland
| | - Tjalf Ziemssen
- Autonomic and Neuroendocrinological Laboratory Dresden, University Hospital Carl Gustav Carus; Technische Universität Dresden, Germany
| | - Walthard Vilser
- Institute of Biomedical Engineering and informatics; Technical University Ilmenau, Germany
- Department of Pediatrics and Adolescent Medicine, Section Neonatalogy; University Hospital, Jena, Germany
| | - Peter M Nilsson
- Department of Clinical Sciences, Lund University; Malmö, Sweden
| | - Martin Magnusson
- Department of Clinical Sciences, Lund University; Malmö, Sweden
- Hypertension in Africa Research Team (HART); North-West University, Potchefstroom, South Africa
- Department of Cardiology; Skåne University Hospital, Malmö, Sweden
- Wallenberg Centre for Molecular Medicine, Lund University; Malmö Sweden
| | - Amra Jujic
- Department of Clinical Sciences, Lund University; Malmö, Sweden
| | - Daniel Mak
- Centre for Regenerative Medicine and Health; Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong, SAR, People's Republic of China
| | - Faans Steyn
- Statistical Consultation Services; North-West University, Potchefstroom, South Africa
| | - Nico T Malan
- Technology Transfer and Innovation-Support Office; Private Bag X1290, North-West University, Potchefstroom 2520, South Africa
| |
Collapse
|
28
|
Bianchetti G, Clementi ME, Sampaolese B, Serantoni C, Abeltino A, De Spirito M, Sasson S, Maulucci G. Metabolic Imaging and Molecular Biology Reveal the Interplay between Lipid Metabolism and DHA-Induced Modulation of Redox Homeostasis in RPE Cells. Antioxidants (Basel) 2023; 12:339. [PMID: 36829896 PMCID: PMC9952658 DOI: 10.3390/antiox12020339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/25/2023] [Accepted: 01/29/2023] [Indexed: 02/04/2023] Open
Abstract
Diabetes-induced oxidative stress induces the development of vascular complications, which are significant causes of morbidity and mortality in diabetic patients. Among these, diabetic retinopathy (DR) is often caused by functional changes in the blood-retinal barrier (BRB) due to harmful oxidative stress events in lipids, proteins, and DNA. Docosahexaenoic acid (DHA) has a potential therapeutic effect against hyperglycemia-induced oxidative damage and apoptotic pathways in the main constituents of BRB, retinal pigment epithelium cells (ARPE-19). Effective antioxidant response elicited by DHA is driven by the activation of the Nrf2/Nqo1 signaling cascade, which leads to the formation of NADH, a reductive agent found in the cytoplasm. Nrf2 also induces the expression of genes encoding enzymes involved in lipid metabolism. This study, therefore, aims at investigating the modulation of lipid metabolism induced by high-glucose (HG) on ARPE-19 cells through the integration of metabolic imaging and molecular biology to provide a comprehensive functional and molecular characterization of the mechanisms activated in the disease, as well the therapeutic role of DHA. This study shows that HG augments RPE metabolic processes by enhancing lipid metabolism, from fatty acid uptake and turnover to lipid biosynthesis and β-oxidation. DHA exerts its beneficial effect by ameliorating lipid metabolism and reducing the increased ROS production under HG conditions. This investigation may provide novel insight for formulating novel treatments for DR by targeting lipid metabolism pathways.
Collapse
Affiliation(s)
- Giada Bianchetti
- Department of Neuroscience, Biophysics Sections, Università Cattolica del Sacro Cuore, Largo Francesco Vito, 1, 00168 Rome, Italy
- Fondazione Policlinico Universitario "A. Gemelli" IRCCS, 00168 Rome, Italy
| | - Maria Elisabetta Clementi
- Institute of Chemical Sciences and Technologies "Giulio Natta" (SCITEC)-CNR, Largo Francesco Vito, 1, 00168 Rome, Italy
| | - Beatrice Sampaolese
- Institute of Chemical Sciences and Technologies "Giulio Natta" (SCITEC)-CNR, Largo Francesco Vito, 1, 00168 Rome, Italy
| | - Cassandra Serantoni
- Department of Neuroscience, Biophysics Sections, Università Cattolica del Sacro Cuore, Largo Francesco Vito, 1, 00168 Rome, Italy
- Fondazione Policlinico Universitario "A. Gemelli" IRCCS, 00168 Rome, Italy
| | - Alessio Abeltino
- Department of Neuroscience, Biophysics Sections, Università Cattolica del Sacro Cuore, Largo Francesco Vito, 1, 00168 Rome, Italy
- Fondazione Policlinico Universitario "A. Gemelli" IRCCS, 00168 Rome, Italy
| | - Marco De Spirito
- Department of Neuroscience, Biophysics Sections, Università Cattolica del Sacro Cuore, Largo Francesco Vito, 1, 00168 Rome, Italy
- Fondazione Policlinico Universitario "A. Gemelli" IRCCS, 00168 Rome, Italy
| | - Shlomo Sasson
- Faculty of Medicine, Institute for Drug Research, The Hebrew University, Jerusalem 911210, Israel
| | - Giuseppe Maulucci
- Department of Neuroscience, Biophysics Sections, Università Cattolica del Sacro Cuore, Largo Francesco Vito, 1, 00168 Rome, Italy
- Fondazione Policlinico Universitario "A. Gemelli" IRCCS, 00168 Rome, Italy
| |
Collapse
|
29
|
Pezzino S, Sofia M, Greco LP, Litrico G, Filippello G, Sarvà I, La Greca G, Latteri S. Microbiome Dysbiosis: A Pathological Mechanism at the Intersection of Obesity and Glaucoma. Int J Mol Sci 2023; 24:ijms24021166. [PMID: 36674680 PMCID: PMC9862076 DOI: 10.3390/ijms24021166] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/03/2023] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
The rate at which obesity is becoming an epidemic in many countries is alarming. Obese individuals have a high risk of developing elevated intraocular pressure and glaucoma. Additionally, glaucoma is a disease of epidemic proportions. It is characterized by neurodegeneration and neuroinflammation with optic neuropathy and the death of retinal ganglion cells (RGC). On the other hand, there is growing interest in microbiome dysbiosis, particularly in the gut, which has been widely acknowledged to play a prominent role in the etiology of metabolic illnesses such as obesity. Recently, studies have begun to highlight the fact that microbiome dysbiosis could play a critical role in the onset and progression of several neurodegenerative diseases, as well as in the development and progression of several ocular disorders. In obese individuals, gut microbiome dysbiosis can induce endotoxemia and systemic inflammation by causing intestinal barrier malfunction. As a result, bacteria and their metabolites could be delivered via the bloodstream or mesenteric lymphatic vessels to ocular regions at the level of the retina and optic nerve, causing tissue degeneration and neuroinflammation. Nowadays, there is preliminary evidence for the existence of brain and intraocular microbiomes. The altered microbiome of the gut could perturb the resident brain-ocular microbiome ecosystem which, in turn, could exacerbate the local inflammation. All these processes, finally, could lead to the death of RGC and neurodegeneration. The purpose of this literature review is to explore the recent evidence on the role of gut microbiome dysbiosis and related inflammation as common mechanisms underlying obesity and glaucoma.
Collapse
Affiliation(s)
- Salvatore Pezzino
- Department of Surgical Sciences and Advanced Technologies “G. F. Ingrassia”, Cannizzaro Hospital, University of Catania, 95126 Catania, Italy
| | - Maria Sofia
- Department of Surgical Sciences and Advanced Technologies “G. F. Ingrassia”, Cannizzaro Hospital, University of Catania, 95126 Catania, Italy
| | - Luigi Piero Greco
- Department of Surgical Sciences and Advanced Technologies “G. F. Ingrassia”, Cannizzaro Hospital, University of Catania, 95126 Catania, Italy
| | - Giorgia Litrico
- Department of Surgical Sciences and Advanced Technologies “G. F. Ingrassia”, Cannizzaro Hospital, University of Catania, 95126 Catania, Italy
| | - Giulia Filippello
- Complex Operative Unit of Ophtalmology, Cannizzaro Hospital, University of Catania, 95126 Catania, Italy
| | - Iacopo Sarvà
- Department of Surgical Sciences and Advanced Technologies “G. F. Ingrassia”, Cannizzaro Hospital, University of Catania, 95126 Catania, Italy
| | - Gaetano La Greca
- Department of Surgical Sciences and Advanced Technologies “G. F. Ingrassia”, Cannizzaro Hospital, University of Catania, 95126 Catania, Italy
| | - Saverio Latteri
- Department of Surgical Sciences and Advanced Technologies “G. F. Ingrassia”, Cannizzaro Hospital, University of Catania, 95126 Catania, Italy
- Correspondence: ; Tel.: +39-0957263584
| |
Collapse
|
30
|
Xiao X, Liu Z, Su G, Liu H, Yin W, Guan Y, Jing S, Du L, Li F, Li N, Yang P. A novel uveitis model induced by lipopolysaccharide in zebrafish. Front Immunol 2022; 13:1042849. [PMID: 36532084 PMCID: PMC9751191 DOI: 10.3389/fimmu.2022.1042849] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 11/18/2022] [Indexed: 12/03/2022] Open
Abstract
Objective Endotoxin-induced uveitis (EIU) is an important tool for human uveitis study. This study was designed to develop a novel EIU model in zebrafish. Methods An EIU model in zebrafish was induced by intravitreal lipopolysaccharide (LPS) injection and was assessed dynamically. Optical coherence tomography (OCT) was used to assess infiltrating cells in the vitreous body. The histological changes wereevaluated using HE staining and immune cells were measured by immunofluorescence. The retinal RNA Sequencing (RNA-Seq) was used to explore the transcriptional changes during inflammation. RNA-Seq data were analyzed using time-course sequencing data analysis (TCseq), ClueGO plugin in Cytoscape, and Gene Set Enrichment Analysis (GSEA) software. Flow cytometry and retinal flat mounts were used to dynamically quantify the immune cells. Results EIU was successfully induced in zebrafish following intravitreal LPS injection. Inflammation appeared at 4 hours post injection (hpi), reached its peak at 24 hpi, and then resolved at 72 hpi. Immunofluorescence confirmed that massive influx ofneutrophils into the iris and vitreous body, and activation of microglia as evidenced by ameboid-shaped appearance in the retina. Retinal RNA-seq during the EIU course identified four gene clusters with distinct expression characteristics related to Toll-likereceptor signaling pathway, cytokine-cytokine receptor interaction, NOD-like receptor signaling pathway, and extracellular matrix (ECM)-receptor interaction, respectively. Prednisone immersion inhibited the inflammatory response of EIU in zebrafish, whichwas confirmed by decreased neutrophils detected in flow cytometry and retinal flat mounts. Conclusions We developed a novel EIU model in zebrafish, which may be particularly useful for gene-editing and high-throughput screening of new drugs for the prevention and treatment of uveitis.
Collapse
Affiliation(s)
- Xiao Xiao
- Henan International Joint Research Laboratory for Ocular Immunology and Retinal Injury Repair, Henan Province Eye Hospital, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhangluxi Liu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology and Chongqing Eye Institute, Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing, China
| | - Guannan Su
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology and Chongqing Eye Institute, Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing, China
| | - Huan Liu
- Henan International Joint Research Laboratory for Ocular Immunology and Retinal Injury Repair, Henan Province Eye Hospital, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wenhui Yin
- Henan International Joint Research Laboratory for Ocular Immunology and Retinal Injury Repair, Henan Province Eye Hospital, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yuxuan Guan
- Henan International Joint Research Laboratory for Ocular Immunology and Retinal Injury Repair, Henan Province Eye Hospital, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shixiang Jing
- Henan International Joint Research Laboratory for Ocular Immunology and Retinal Injury Repair, Henan Province Eye Hospital, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Liping Du
- Henan International Joint Research Laboratory for Ocular Immunology and Retinal Injury Repair, Henan Province Eye Hospital, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Fuzhen Li
- Henan International Joint Research Laboratory for Ocular Immunology and Retinal Injury Repair, Henan Province Eye Hospital, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Na Li
- Henan International Joint Research Laboratory for Ocular Immunology and Retinal Injury Repair, Henan Province Eye Hospital, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Peizeng Yang
- Henan International Joint Research Laboratory for Ocular Immunology and Retinal Injury Repair, Henan Province Eye Hospital, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China,The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology and Chongqing Eye Institute, Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing, China,*Correspondence: Peizeng Yang,
| |
Collapse
|
31
|
Wang N, Yang Y, Liu Y, Huang L, Gu M, Wu Y, Xu L, Sun H, Guo W. Magnolol limits NFκB-dependent inflammation by targeting PPARγ relieving retinal ischemia/reperfusion injury. Int Immunopharmacol 2022; 112:109242. [PMID: 36152538 DOI: 10.1016/j.intimp.2022.109242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/29/2022] [Accepted: 09/06/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND Glaucoma is the leading cause of irreversible blindness in the world. Elevated intraocular pressure (IOP) is recognized as one of the most critical factors, but the loss of retinal ganglia cells (RGCs) often persists when IOP is controlled. Recently, a large number of studies focus on the inflammatory and immune responses in the occurrence and development of glaucoma. Magnolol (MAG), the principal ingredient of magnoliae officinalis cortex, has anti-inflammatory effects, but its role and mechanism in retinal protection need to be further studied. METHODS The neurodegeneration of retina in mice model following ischemia/reperfusion (IR) injury was evaluated by immunohistochemistry, hematoxylin and eosin (H&E) staining and electroretinography (ERG). The inflammation-regulatory effect of MAG was detected by quantitative RT-PCR, western blot, and immunohistochemistry. Peroxisome proliferator-activated receptor-γ (PPARγ) inhibitor assays by H&E staining and western blot were used to test the target and mechanism pathway of MAG. RESULTS We found MAG relieved IR-induced retinal damages and inflammation. Further studies revealed MAG alleviated nuclear factor kappa B (NFκB)-dependent inflammatory process by preserving the expression of NFκB inhibitor alpha (IκBα), and it modulated microglia polarization after IR injury. PPARγ was a primary target of MAG, and treatment with PPARγ inhibitor GW9662 attenuated the neuroprotective and anti-inflammatory effects of MAG. CONCLUSIONS Our findings revealed that MAG inhibits NFκB-dependent inflammatory processes by elevating PPARγ in mice retinas to achieve its neuroprotective role following IR, which suggesting that MAG could be developed to a novel anti-inflammatory therapeutic agent for relieving the progression of glaucoma.
Collapse
Affiliation(s)
- Ning Wang
- Department of Ophthalmology, Shanghai 9th People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200011, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Yijie Yang
- Department of Ophthalmology, Shanghai 9th People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200011, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Yixin Liu
- Department of Ophthalmology, Shanghai 9th People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200011, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Lulu Huang
- Department of Ophthalmology, Shanghai 9th People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200011, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Mengyang Gu
- Department of Ophthalmology, Shanghai 9th People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200011, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Yue Wu
- Department of Ophthalmology, Shanghai 9th People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200011, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Li Xu
- Department of Ophthalmology, Shanghai 9th People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200011, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China.
| | - Hao Sun
- Department of Ophthalmology, Shanghai 9th People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200011, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China.
| | - Wenyi Guo
- Department of Ophthalmology, Shanghai 9th People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200011, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China.
| |
Collapse
|
32
|
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
|
33
|
Investigation of DHA-Induced Regulation of Redox Homeostasis in Retinal Pigment Epithelium Cells through the Combination of Metabolic Imaging and Molecular Biology. Antioxidants (Basel) 2022; 11:antiox11061072. [PMID: 35739970 PMCID: PMC9219962 DOI: 10.3390/antiox11061072] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 05/23/2022] [Accepted: 05/26/2022] [Indexed: 12/13/2022] Open
Abstract
Diabetes-induced oxidative stress leads to the onset of vascular complications, which are major causes of disability and death in diabetic patients. Among these, diabetic retinopathy (DR) often arises from functional alterations of the blood-retinal barrier (BRB) due to damaging oxidative stress reactions in lipids, proteins, and DNA. This study aimed to investigate the impact of the ω3-polyunsaturated docosahexaenoic acid (DHA) on the regulation of redox homeostasis in the human retinal pigment epithelial (RPE) cell line (ARPE-19) under hyperglycemic-like conditions. The present results show that the treatment with DHA under high-glucose conditions activated erythroid 2-related factor Nrf2, which orchestrates the activation of cellular antioxidant pathways and ultimately inhibits apoptosis. This process was accompanied by a marked increase in the expression of NADH (Nicotinamide Adenine Dinucleotide plus Hydrogen) Quinone Oxidoreductase 1 (Nqo1), which is correlated with a contextual modulation and intracellular re-organization of the NAD+/NADH redox balance. This investigation of the mechanisms underlying the impairment induced by high levels of glucose on redox homeostasis of the BRB and the subsequent recovery provided by DHA provides both a powerful indicator for the detection of RPE cell impairment as well as a potential metabolic therapeutic target for the early intervention in its treatment.
Collapse
|
34
|
Wang S, Yu Q, Wang Y, Xu C, Niu G, Liu R. CircSLC16A12 absence inhibits high glucose-induced dysfunction in retinal microvascular endothelial cells through mediating miR-140-3p/FGF2 axis in diabetic retinopathy. Curr Eye Res 2022; 47:759-769. [PMID: 35179428 DOI: 10.1080/02713683.2022.2025845] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND Diabetic retinopathy (DR) is a common microvascular complication of diabetes mellitus which can cause irreversible visual impairment and blindness. We intended to investigate the function of circular RNA (circRNA) solute carrier family 16 member 12 (SLC16A12) in DR progression. METHODS Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and Western blot assay were applied to measure RNA and protein expression. Cell apoptosis was analyzed by flow cytometry (FCM) analysis. The angiogenesis ability was assessed by tube formation assay. Enzyme-linked immunosorbent assay (ELISA) was performed to analyze the release of inflammatory cytokines. Cell oxidative stress status was evaluated using commercial kits. Dual-luciferase reporter assay, RNA immunoprecipitation (RIP) assay, and RNA-pull down assay were conducted to confirm the intermolecular interactions. RESULTS CircSLC16A12 level was enhanced in the serum samples of DR patients and high glucose (HG)-treated HRECs. CircSLC16A12 absence protected HRECs from HG-induced apoptosis, blood-retinal barrier (BRB) injury, tube formation, inflammatory response, and oxidative stress. CircSLC16A12 acted as a sponge for microRNA-140-3p (miR-140-3p), and circSLC16A12 knockdown-mediated effects were largely reversed by the absence of miR-140-3p in HRECs under HG condition. miR-140-3p interacted with the 3' untranslated region (3'UTR) of fibroblast growth factor 2 (FGF2), and the overexpression of FGF2 largely overturned miR-140-3p overexpression-mediated effects in HRECs. CircSLC16A12 interference reduced the expression of FGF2 by up-regulating miR-140-3p in HRECs. CONCLUSION CircSLC16A12 silencing suppressed HG-induced dysfunction in HRECs partly by targeting miR-140-3p/FGF2 axis.
Collapse
Affiliation(s)
- Shanshan Wang
- Department of Ophthalmic Clinic, Harbin Eye Hospital, Harbin, 150000, Heilongjiang, China
| | - Qing Yu
- Department of Eye Care, Harbin Eye Hospital, Harbin, 150000, Heilongjiang, China
| | - Yujue Wang
- Department of Ophthalmic Clinic, Harbin Eye Hospital, Harbin, 150000, Heilongjiang, China
| | - Chunyue Xu
- Department of Ophthalmic Clinic, Harbin Eye Hospital, Harbin, 150000, Heilongjiang, China
| | - Guoxiang Niu
- Department of Eye Care, Harbin Eye Hospital, Harbin, 150000, Heilongjiang, China
| | - Rui Liu
- Department of Ophthalmic Clinic, Harbin Eye Hospital, Harbin, 150000, Heilongjiang, China
| |
Collapse
|
35
|
Wang L, Wei X. T Cell-Mediated Autoimmunity in Glaucoma Neurodegeneration. Front Immunol 2022; 12:803485. [PMID: 34975917 PMCID: PMC8716691 DOI: 10.3389/fimmu.2021.803485] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 12/01/2021] [Indexed: 02/05/2023] Open
Abstract
Glaucoma as the leading neurodegenerative disease leads to blindness in 3.6 million people aged 50 years and older worldwide. For many decades, glaucoma therapy has primarily focused on controlling intraocular pressure (IOP) and sound evidence supports its role in delaying the progress of retinal ganglial cell (RGC) damage and protecting patients from vision loss. Meanwhile, accumulating data point to the immune-mediated attack of the neural retina as the underlying pathological process behind glaucoma that may come independent of raised IOP. Recently, some scholars have suggested autoimmune aspects in glaucoma, with autoreactive T cells mediating the chief pathogenic process. This autoimmune process, as well as the pathological features of glaucoma, largely overlaps with other neurodegenerative diseases in the central nervous system (CNS), including Alzheimer’s disease, Parkinson’s disease, and multiple sclerosis. In addition, immune modulation therapy, which is regarded as a potential solution for glaucoma, has been boosted in trials in some CNS neurodegenerative diseases. Thus, novel insights into the T cell-mediated immunity and treatment in CNS neurodegenerative diseases may serve as valuable inspirations for ophthalmologists. This review focuses on the role of T cell-mediated immunity in the pathogenesis of glaucoma and discusses potential applications of relevant findings of CNS neurodegenerative diseases in future glaucoma research.
Collapse
Affiliation(s)
- Lixiang Wang
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| | - Xin Wei
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China.,Department of Ophthalmology, Shangjin Nanfu Hospital, Chengdu, China
| |
Collapse
|
36
|
Martínez-Alberquilla I, Gasull X, Pérez-Luna P, Seco-Mera R, Ruiz-Alcocer J, Crooke A. Neutrophils and neutrophil extracellular trap components: Emerging biomarkers and therapeutic targets for age-related eye diseases. Ageing Res Rev 2022; 74:101553. [PMID: 34971794 DOI: 10.1016/j.arr.2021.101553] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 11/17/2021] [Accepted: 12/23/2021] [Indexed: 12/14/2022]
Abstract
Age-related eye diseases, including dry eye, glaucoma, age-related macular degeneration, and diabetic retinopathy, represent a major global health issue based on their increasing prevalence and disabling action. Unraveling the molecular mechanisms underlying these diseases will provide novel opportunities to reduce the burden of age-related eye diseases and improve eye health, contributing to sustainable development goals achievement. The impairment of neutrophil extracellular traps formation/degradation processes seems to be one of these mechanisms. These traps formed by a meshwork of DNA and neutrophil cytosolic granule proteins may exacerbate the inflammatory response promoting chronic inflammation, a pivotal cause of age-related diseases. In this review, we describe current findings that suggest the role of neutrophils and their traps in the pathogenesis of the above-mentioned age-related eye diseases. Furthermore, we discuss why these cells and their constituents could be biomarkers and therapeutic targets for dry eye, glaucoma, age-related macular degeneration, and diabetic retinopathy. We also examine the therapeutic potential of some neutrophil function modulators and provide several recommendations for future research in age-related eye diseases.
Collapse
Affiliation(s)
- Irene Martínez-Alberquilla
- Department of Optometry and Vision, Faculty of Optics and Optometry, Universidad Complutense de Madrid, Madrid, Spain; Clinical and Experimental Eye Research Group, UCM 971009, Faculty of Optics and Optometry, Universidad Complutense de Madrid, Madrid, Spain
| | - Xavier Gasull
- Neurophysiology Laboratory, Department of Biomedicine, Medical School, Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Patricia Pérez-Luna
- Department of Biochemistry and Molecular Biology, Faculty of Optics and Optometry, Universidad Complutense de Madrid, Madrid, Spain
| | - Rubén Seco-Mera
- Department of Biochemistry and Molecular Biology, Faculty of Optics and Optometry, Universidad Complutense de Madrid, Madrid, Spain
| | - Javier Ruiz-Alcocer
- Department of Optometry and Vision, Faculty of Optics and Optometry, Universidad Complutense de Madrid, Madrid, Spain; Clinical and Experimental Eye Research Group, UCM 971009, Faculty of Optics and Optometry, Universidad Complutense de Madrid, Madrid, Spain
| | - Almudena Crooke
- Department of Biochemistry and Molecular Biology, Faculty of Optics and Optometry, Universidad Complutense de Madrid, Madrid, Spain; Clinical and Experimental Eye Research Group, UCM 971009, Faculty of Optics and Optometry, Universidad Complutense de Madrid, Madrid, Spain.
| |
Collapse
|
37
|
Rudraraju M, Narayanan SP, Somanath PR. Distinct Mechanisms of Human Retinal Endothelial Barrier Modulation In Vitro by Mediators of Diabetes and Uveitis. Life (Basel) 2021; 12:life12010033. [PMID: 35054426 PMCID: PMC8779223 DOI: 10.3390/life12010033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 11/30/2021] [Accepted: 12/10/2021] [Indexed: 12/04/2022] Open
Abstract
Ocular diseases such as diabetic retinopathy (DR) and uveitis are associated with injury to the blood–retinal barrier (BRB). Whereas high glucose (HG) and advanced glycation end products (AGE) contribute to DR, bacterial infections causing uveitis are triggered by endotoxins such as lipopolysaccharide (LPS). It is unclear how HG, AGE, and LPS affect human retinal endothelial cell (HREC) junctions. Moreover, tumor necrosis factor-α (TNFα) is elevated in both DR and ocular infections. In the current study, we determined the direct effects of HG, AGE, TNFα, and LPS on the expression and intracellular distribution of claudin-5, VE-cadherin, and β-catenin in HRECs and how these mediators affect Akt and P38 MAP kinase that have been implicated in ocular pathologies. In our results, whereas HG, AGE, and TNFα activated both Akt and P38 MAPK, LPS treatment suppressed Akt but increased P38 MAPK phosphorylation. Furthermore, while treatment with AGE and HG increased cell-junction protein expression in HRECs, LPS elicited a paradoxical effect. By contrast, when HG treatment increased HREC-barrier resistance, AGE and LPS stimulation compromised it, and TNFα had no effect. Together, our results demonstrated the differential effects of the mediators of diabetes and infection on HREC-barrier modulation leading to BRB injury.
Collapse
Affiliation(s)
- Madhuri Rudraraju
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA 30912, USA;
- Research Division, Charlie Norwood VA Medical Center, Augusta, GA 30912, USA
| | - S. Priya Narayanan
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA 30912, USA;
- Research Division, Charlie Norwood VA Medical Center, Augusta, GA 30912, USA
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
- Vision Discovery Institute, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
- Correspondence: (S.P.N.); (P.R.S.); Tel.: +1-706-721-4250 (P.R.S.)
| | - Payaningal R. Somanath
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA 30912, USA;
- Research Division, Charlie Norwood VA Medical Center, Augusta, GA 30912, USA
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
- Correspondence: (S.P.N.); (P.R.S.); Tel.: +1-706-721-4250 (P.R.S.)
| |
Collapse
|
38
|
Xu M, Jiang Y, Su L, Chen X, Shao X, Ea V, Shang Z, Zhang X, Barnstable CJ, Li X, Tombran-Tink J. Novel Regulators of Retina Neovascularization: A Proteomics Approach. J Proteome Res 2021; 21:101-117. [PMID: 34919406 DOI: 10.1021/acs.jproteome.1c00547] [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] [Indexed: 12/12/2022]
Abstract
The purpose of this study was to identify proteins that regulate vascular remodeling in an ROP mouse model. Pups were subjected to fluctuating oxygen levels and retinas sampled during vessel regression (PN12) or neovascularization (PN17) for comparative SWATH-MS proteomics using liquid chromatography-tandem mass spectrometry (LC-MS/MS). We developed a human retinal endothelial cell (HREC) ROP correlate to validate the expression of retina neovascular-specific markers. A total of 5191 proteins were identified in OIR retinas with 498 significantly regulated in elevated oxygen and 345 after a return to normoxia. A total of 122 proteins were uniquely regulated during vessel regression and 69 during neovascularization (FC ≥ 1.5; p ≤ 0.05), with several validated by western blot analyses. Expressions of 56/69 neovascular-specific proteins were confirmed in hypoxic HRECs with 23 regulated in the same direction as OIR neovascular retinas. These proteins control angiogenesis-related processes including matrix remodeling, cell migration, adhesion, and proliferation. RNAi and transfection overexpression studies confirmed that VASP and ECH1, showing the highest levels in hypoxic HRECs, promoted human umbilical vein (HUVEC) and HREC cell proliferation, while SNX1 and CD109, showing the lowest levels, inhibited their proliferation. These proteins are potential biomarkers and exploitable intervention tools for vascular-related disorders. The proteomics data set generated has been deposited to the ProteomeXchange/iProX Consortium with the Identifier:PXD029208.
Collapse
Affiliation(s)
- Manhong Xu
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin 300384, China
| | - Yilin Jiang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin 300384, China
| | - Lin Su
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin 300384, China
| | - Xin Chen
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin 300384, China
| | - Xianfeng Shao
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Lifeomics, National Center for Protein Sciences (The PHOENIX Center, Beijing), Beijing 102206, China
| | - Vicki Ea
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin 300384, China
| | - Zhenying Shang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin 300384, China
| | - Xiaomin Zhang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin 300384, China
| | - Colin J Barnstable
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin 300384, China.,Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, Pennsylvania 17033-0850, United States
| | - Xiaorong Li
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin 300384, China
| | - Joyce Tombran-Tink
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin 300384, China.,Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, Pennsylvania 17033-0850, United States
| |
Collapse
|
39
|
Vernazza S, Oddone F, Tirendi S, Bassi AM. Risk Factors for Retinal Ganglion Cell Distress in Glaucoma and Neuroprotective Potential Intervention. Int J Mol Sci 2021; 22:7994. [PMID: 34360760 PMCID: PMC8346985 DOI: 10.3390/ijms22157994] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 07/21/2021] [Accepted: 07/23/2021] [Indexed: 12/14/2022] Open
Abstract
Retinal ganglion cells (RGCs) are a population of neurons of the central nervous system (CNS) extending with their soma to the inner retina and with their axons to the optic nerve. Glaucoma represents a group of neurodegenerative diseases where the slow progressive death of RGCs results in a permanent loss of vision. To date, although Intra Ocular Pressure (IOP) is considered the main therapeutic target, the precise mechanisms by which RGCs die in glaucoma have not yet been clarified. In fact, Primary Open Angle Glaucoma (POAG), which is the most common glaucoma form, also occurs without elevated IOP. This present review provides a summary of some pathological conditions, i.e., axonal transport blockade, glutamate excitotoxicity and changes in pro-inflammatory cytokines along the RGC projection, all involved in the glaucoma cascade. Moreover, neuro-protective therapeutic approaches, which aim to improve RGC degeneration, have also been taken into consideration.
Collapse
Affiliation(s)
- Stefania Vernazza
- Department of Experimental Medicine (DIMES), University of Genoa, 16126 Genoa, Italy; (S.T.); (A.M.B.)
| | | | - Sara Tirendi
- Department of Experimental Medicine (DIMES), University of Genoa, 16126 Genoa, Italy; (S.T.); (A.M.B.)
- Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research (Centro 3R), 56122 Pisa, Italy
| | - Anna Maria Bassi
- Department of Experimental Medicine (DIMES), University of Genoa, 16126 Genoa, Italy; (S.T.); (A.M.B.)
- Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research (Centro 3R), 56122 Pisa, Italy
| |
Collapse
|
40
|
Byrne EM, Llorián-Salvador M, Tang M, Margariti A, Chen M, Xu H. IL-17A Damages the Blood-Retinal Barrier through Activating the Janus Kinase 1 Pathway. Biomedicines 2021; 9:831. [PMID: 34356895 PMCID: PMC8301352 DOI: 10.3390/biomedicines9070831] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 07/06/2021] [Accepted: 07/14/2021] [Indexed: 12/16/2022] Open
Abstract
Blood-retinal barrier (BRB) dysfunction underlies macular oedema in many sight-threatening conditions, including diabetic macular oedema, neovascular age-related macular degeneration and uveoretinitis. Inflammation plays an important role in BRB dysfunction. This study aimed to understand the role of the inflammatory cytokine IL-17A in BRB dysfunction and the mechanism involved. Human retinal pigment epithelial (RPE) cell line ARPE19 and murine brain endothelial line bEnd.3 were cultured on transwell membranes to model the outer BRB and inner BRB, respectively. IL-17A treatment (3 days in bEnd.3 cells and 6 days in ARPE19 cells) disrupted the distribution of claudin-5 in bEnd.3 cells and ZO-1 in ARPE19 cells, reduced the transepithelial/transendothelial electrical resistance (TEER) and increased permeability to FITC-tracers in vitro. Intravitreal (20 ng/1 μL/eye) or intravenous (20 ng/g) injection of recombinant IL-17A induced retinal albumin leakage within 48 h in C57BL/6J mice. Mechanistically, IL-17A induced Janus kinase 1 (JAK1) phosphorylation in bEnd.3 but not ARPE19 cells. Blocking JAK1 with Tofacitinib prevented IL-17A-mediated claudin-5 dysmorphia in bEnd.3 cells and reduced albumin leakage in IL-17A-treated mice. Our results suggest that IL-17A can damage the BRB through the activating the JAK1 signaling pathway, and targeting this pathway may be a novel approach to treat inflammation-induced macular oedema.
Collapse
Affiliation(s)
| | | | | | | | | | - Heping Xu
- The Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, Belfast BT9 7BL, UK; (E.M.B.); (M.L.-S.); (M.T.); (A.M.); (M.C.)
| |
Collapse
|
41
|
Napoli D, Biagioni M, Billeri F, Di Marco B, Orsini N, Novelli E, Strettoi E. Retinal Pigment Epithelium Remodeling in Mouse Models of Retinitis Pigmentosa. Int J Mol Sci 2021; 22:ijms22105381. [PMID: 34065385 PMCID: PMC8161377 DOI: 10.3390/ijms22105381] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/13/2021] [Accepted: 05/17/2021] [Indexed: 12/28/2022] Open
Abstract
In retinitis pigmentosa (RP), one of many possible genetic mutations causes rod degeneration, followed by cone secondary death leading to blindness. Accumulating evidence indicates that rod death triggers multiple, non-cell-autonomous processes, which include oxidative stress and inflammation/immune responses, all contributing to cone demise. Inflammation relies on local microglia and recruitment of immune cells, reaching the retina through breakdowns of the inner blood retinal barrier (iBRB). Leakage in the inner retina vasculature suggests similarly altered outer BRB, formed by junctions between retinal pigment epithelium (RPE) cells, which are crucial for retinal homeostasis, immune response, and privilege. We investigated the RPE structural integrity in three models of RP (rd9, rd10, and Tvrm4 mice) by immunostaining for zonula occludens-1 (ZO-1), an essential regulatory component of tight junctions. Quantitative image analysis demonstrated discontinuities in ZO-1 profiles in all mutants, despite different degrees of photoreceptor loss. ZO-1 interruption zones corresponded to leakage of in vivo administered, fluorescent dextran through the choroid-RPE interface, demonstrating barrier dysfunction. Dexamethasone, administered to rd10 mice for rescuing cones, also rescued RPE structure. Thus, previously undetected, stereotyped abnormalities occur in the RPE of RP mice; pharmacological targeting of inflammation supports a feedback loop leading to simultaneous protection of cones and the RPE.
Collapse
Affiliation(s)
- Debora Napoli
- CNR Neuroscience Institute, 56124 Pisa, Italy; (M.B.); (F.B.); (B.D.M.); (N.O.); (E.N.)
- Correspondence: (D.N.); (E.S.); Tel.: +39-0503153157 (E.S.)
| | - Martina Biagioni
- CNR Neuroscience Institute, 56124 Pisa, Italy; (M.B.); (F.B.); (B.D.M.); (N.O.); (E.N.)
| | - Federico Billeri
- CNR Neuroscience Institute, 56124 Pisa, Italy; (M.B.); (F.B.); (B.D.M.); (N.O.); (E.N.)
- Department of Biology, University of Pisa, 56126 Pisa, Italy
| | - Beatrice Di Marco
- CNR Neuroscience Institute, 56124 Pisa, Italy; (M.B.); (F.B.); (B.D.M.); (N.O.); (E.N.)
| | - Noemi Orsini
- CNR Neuroscience Institute, 56124 Pisa, Italy; (M.B.); (F.B.); (B.D.M.); (N.O.); (E.N.)
- Regional Doctorate School in Neuroscience, Universities of Florence, Pisa and Siena, 50139 Florence, Italy
| | - Elena Novelli
- CNR Neuroscience Institute, 56124 Pisa, Italy; (M.B.); (F.B.); (B.D.M.); (N.O.); (E.N.)
| | - Enrica Strettoi
- CNR Neuroscience Institute, 56124 Pisa, Italy; (M.B.); (F.B.); (B.D.M.); (N.O.); (E.N.)
- Correspondence: (D.N.); (E.S.); Tel.: +39-0503153157 (E.S.)
| |
Collapse
|