1
|
Wu KY, Dave A, Nirwal GK, Giunta M, Nguyen VDH, Tran SD. Exosome Innovations in Ophthalmology and Sjögren's Syndrome. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025. [PMID: 40360847 DOI: 10.1007/5584_2025_865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
Exosomes, a subset of extracellular vesicles, have emerged as potential therapeutic agents in ophthalmology due to their ability to modulate immune responses, facilitate cellular communication, and promote tissue repair. This chapter explores the potential applications of exosome-based therapies in corneal and anterior segment disorders, retinal diseases, glaucoma, and Sjögren's syndrome. In corneal disorders, mesenchymal stem cell (MSC)-derived secretomes have shown promise in accelerating wound healing, reducing fibrosis, and modulating inflammation, with hydrogel encapsulation strategies potentially enhancing their efficacy. In retinal diseases, exosomes may provide neuroprotective effects in age-related macular degeneration, diabetic retinopathy, and retinitis pigmentosa by modulating oxidative stress and inflammation. In glaucoma, secretome-based therapies could support retinal ganglion cell survival and optic nerve regeneration, though their impact on intraocular pressure via the trabecular meshwork remains uncertain. Additionally, exosomal biomarkers in aqueous humor are being investigated as potential diagnostic tools. In Sjögren's syndrome, exosomal biomarkers may facilitate earlier detection, while stem cell-derived exosomes hold promise in modulating immune responses and restoring glandular function. Despite encouraging preclinical and early clinical findings, standardization, scalability, and long-term safety must be addressed before clinical translation. Future research will focus on optimizing exosome-based therapies and exploring their feasibility for ophthalmic applications.
Collapse
Affiliation(s)
- Kevin Y Wu
- Department of Surgery, Division of Ophthalmology, University of Sherbrooke, Sherbrooke, QC, Canada.
| | - Archan Dave
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Gurleen K Nirwal
- Department of Zoology, University of British Columbia, Vancouver, BC, Canada
| | - Michel Giunta
- Department of Surgery, Division of Ophthalmology, University of Sherbrooke, Sherbrooke, QC, Canada
| | | | - Simon D Tran
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC, Canada
| |
Collapse
|
2
|
Wu D, Fan Z, Hu Y, Chen Y, Tian R, Wang C, He H, Yang Y, Zhang G. Identifying potential tear biomarkers in premature infants with retinopathy of prematurity based on proteome and transcriptome analysis. Graefes Arch Clin Exp Ophthalmol 2025:10.1007/s00417-025-06838-1. [PMID: 40343554 DOI: 10.1007/s00417-025-06838-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 02/26/2025] [Accepted: 04/21/2025] [Indexed: 05/11/2025] Open
Abstract
AIM To identify the potential tear fluid biomarkers in premature infants with and without retinopathy of prematurity (ROP) based on proteomic and transcriptomic analysis. METHODS Tears were collected from the 46 eyes of the 23 enrolled premature infants, with and without ROP. Data-independent acquisition (DIA) mass spectrometry was utilized for the quantitative proteomic analysis of the two groups. Two published transcriptome datasets involving mouse oxygen-induced retinopathy (OIR) model data were selected from the Gene Expression Omnibus (GEO) database. iDEP (integrated Differential Expression and Pathway analysis) were used for differential expression analysis. Gene Ontology (GO)-based functional and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis were performed. RESULTS In this study, a total of 1742 proteins were quantified from the two groups. 55 differentially expressed proteins closely related to immune and angiogenesis processes were identified, including 33 highly expressed as well as 22 lowly expressed in the ROP group. Combined with RNA-seq data from OIR model, we screened two particularly critical proteins, LYN and filamin A (FLNA), which were both expressed at significantly elevated levels. CONCLUSIONS According to the findings of the tear proteomics data, we hypothesized two particularly critical proteins, LYN and FLNA, may serve as pivotal regulators of immune and angiogenesis processes in ROP. These results will assist in the provision of new potential targets for the diagnosis of ROP.
Collapse
Affiliation(s)
- Dongting Wu
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Zixin Fan
- Shenzhen Eye Hospital, Shenzhen Eye Medical Center, Southern Medical University, Shenzhen, 518040, Guangdong, China
| | - Yarou Hu
- Shenzhen Eye Hospital, Shenzhen Eye Medical Center, Southern Medical University, Shenzhen, 518040, Guangdong, China
| | - Yi Chen
- Shenzhen Eye Hospital, Shenzhen Eye Medical Center, Southern Medical University, Shenzhen, 518040, Guangdong, China
| | - Ruyin Tian
- Shenzhen Eye Hospital, Shenzhen Eye Medical Center, Southern Medical University, Shenzhen, 518040, Guangdong, China
| | - Cui Wang
- Shenzhen Eye Hospital, Shenzhen Eye Medical Center, Southern Medical University, Shenzhen, 518040, Guangdong, China
| | - Honghui He
- Shenzhen Eye Hospital, Shenzhen Eye Medical Center, Southern Medical University, Shenzhen, 518040, Guangdong, China
| | - Yuhang Yang
- Shenzhen Eye Hospital, Shenzhen Eye Medical Center, Southern Medical University, Shenzhen, 518040, Guangdong, China
| | - Guoming Zhang
- Shenzhen Eye Hospital, Shenzhen Eye Medical Center, Southern Medical University, Shenzhen, 518040, Guangdong, China.
| |
Collapse
|
3
|
Li W, Zhang Y, Zhu H, Su N, Sun R, Mao X, Yang Q, Yuan S. CAVIN3 deficiency promotes vascular normalization in ocular neovascular disease via ERK/JAG1 signaling pathway. JCI Insight 2025; 10:e187836. [PMID: 40337864 DOI: 10.1172/jci.insight.187836] [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: 10/10/2024] [Accepted: 03/27/2025] [Indexed: 05/09/2025] Open
Abstract
Multiple members of the caveolae-associated protein (Cavin) family are implicated in angiogenesis. However, the specific role of CAVIN3 in pathological angiogenesis within the eye remains unclear. The present study demonstrated that CAVIN3 knockdown in endothelial cells (ECs) promoted vascular normalization in ocular pathological neovascularization. Elevated CAVIN3 expression was observed in the ECs of retinal pigment epithelium/choroid complexes from patients with neovascular age-related macular degeneration and fibrovascular membranes from patients with proliferative diabetic retinopathy. Additionally, upregulated Cavin3 expression was detected in laser-induced choroidal neovascularization (CNV) and oxygen-induced retinopathy (OIR) mouse models. In both OIR and CNV mice, Cavin3 knockdown inhibited pathological neovascularization. Cavin3 deficiency further disrupted EC proliferation and vascular sprouting, thereby promoting vascular normalization by partially restoring microenvironmental hypoxia and reestablishing pericyte-EC interactions. Mechanistically, we demonstrated that zinc finger E-box-binding homeobox 1 (ZEB1) regulated CAVIN3 transcription in ECs under hypoxic conditions. CAVIN3 deficiency modulated pathological vascularization by inhibiting ERK phosphorylation, which downregulated jagged 1 (JAG1) expression. Conclusively, this study elucidated the protective role of endothelial CAVIN3 deficiency in pathological neovascularization models, addressing a gap in understanding the regulatory role of Cavins in angiogenesis. These findings suggested a therapeutic direction for ocular neovascular diseases.
Collapse
Affiliation(s)
- Weiqi Li
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yeran Zhang
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hongjing Zhu
- Department of Ophthalmology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Na Su
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ruxu Sun
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiying Mao
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qin Yang
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Songtao Yuan
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
4
|
Zhang S, Liu J, Zhao H, Gao Y, Ren C, Zhang X. What do You Need to Know after Diabetes and before Diabetic Retinopathy? Aging Dis 2025:AD.2025.0289. [PMID: 40354381 DOI: 10.14336/ad.2025.0289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Accepted: 04/30/2025] [Indexed: 05/14/2025] Open
Abstract
Diabetic retinopathy (DR) is a leading cause of vision impairment and blindness among individuals with diabetes mellitus. Current clinical diagnostic criteria mainly base on visible vascular structure changes, which are insufficient to identify diabetic patients without clinical DR (NDR) but with dysfunctional retinopathy. This review focuses on retinal endothelial cells (RECs), the first cells to sense and respond to elevated blood glucose. As blood glucose rises, RECs undergo compensatory and transitional phases, and the correspondingly altered molecules are likely to become biomarkers and targets for early prediction and treatment of NDR with dysfunctional retinopathy. This article elaborated the possible pathophysiological processes focusing on RECs and summarized recently published and reliable biomarkers for early screening and emerging intervention strategies for NDR patients with dysfunctional retinopathy. Additionally, references for clinical medication selection and lifestyle recommendations for this population are provided. This review aims to deepen the understanding of REC biology and NDR pathophysiology, emphasizes the importance of early detection and intervention, and points out future directions to improve the diagnosis and treatment of NDR with dysfunctional retinopathy and to reduce the occurrence of DR.
Collapse
Affiliation(s)
- Shiyu Zhang
- Department of Ophthalmology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jia Liu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Laboratory for Clinical Medicine, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Capital Medical University, Beijing, China
| | - Heng Zhao
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Laboratory for Clinical Medicine, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Capital Medical University, Beijing, China
| | - Yuan Gao
- Department of Ophthalmology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Changhong Ren
- Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Center of Stroke, Beijing Institute of Brain Disorder, Capital Medical University, Beijing, China
| | - Xuxiang Zhang
- Department of Ophthalmology, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
5
|
Klier S, Dananberg J, Masaki L, Bhisitkul RB, Khanani AM, Maturi R, Salehi-Had H, Mallinckrodt CH, Rathmell JM, Ghosh A, Sapieha P. Safety and Efficacy of Senolytic UBX1325 in Diabetic Macular Edema. NEJM EVIDENCE 2025; 4:EVIDoa2400009. [PMID: 40261111 DOI: 10.1056/evidoa2400009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
BACKGROUND We tested the ability of a single intravitreal injection of foselutoclax (hereafter UBX1325), a novel senolytic small molecule inhibitor of antiapoptotic protein B-cell lymphoma-extra large, to mitigate the impact of diabetic macular edema. METHODS Patients with diabetic macular edema with prior suboptimal response to anti-vascular endothelial growth factor treatment were randomly assigned (1:1) to either a single intravitreal injection of 10 μg of UBX1325 or sham and were followed for up to 48 weeks. The primary trial objective was to evaluate the safety and side-effect profile of UBX1325 as assessed by ocular and systemic treatment-emergent adverse events (TEAEs). Our secondary objective was to probe efficacy, defined as mean changes from baseline for UBX1325 versus sham in best corrected visual acuity measured in Early Treatment of Diabetic Retinopathy Study (ETDRS) letters (range, 0-100 letters, higher scores indicate better vision) and retinal structure. RESULTS Between June 2021 and April 2022, 65 participants (32.3% women) were randomly assigned to either UBX1325 (n=32) or sham (n=33). There were four TEAEs of Grade 3 or greater in the sham group, of which three were considered serious, while there were five in the UBX1325 group of Grade 3 or greater and considered serious. There were no apparent between-group differences with respect to vital signs, electrocardiograms, or routine blood chemistries. For the secondary outcome of efficacy, the difference between UBX1325 and sham in mean change to week 48 in best corrected visual acuity was 5.6 more ETDRS letters (95% confidence interval, -1.5 to 12.7). CONCLUSIONS In this sham-controlled trial there were no TEAEs that led to discontinuation of treatment with UBX1325 compared with sham. There were trends suggestive of potential efficacy; larger trials are needed to further evaluate these findings. (Funded by UNITY Biotechnology; ClinicalTrials.gov number, NCT04857996.).
Collapse
Affiliation(s)
| | | | | | | | - Arshad M Khanani
- Sierra Eye Associates, Reno, NV, and Reno School of Medicine, The University of Nevada, Reno, NV
| | - Raj Maturi
- Midwest Eye Institute, Indianapolis
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis
| | - Hani Salehi-Had
- Retina Associates of Southern California, Huntington Beach, CA
| | | | | | | | - Przemyslaw Sapieha
- UNITY Biotechnology, San Francisco
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal
| |
Collapse
|
6
|
Liang C, Liu L, Yu W, Shi Q, Zheng J, Lyu J, Zhong J. Construction and validation of risk prediction models for different subtypes of retinal vein occlusion. ADVANCES IN OPHTHALMOLOGY PRACTICE AND RESEARCH 2025; 5:107-116. [PMID: 40230508 PMCID: PMC11995075 DOI: 10.1016/j.aopr.2025.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 03/02/2025] [Accepted: 03/13/2025] [Indexed: 04/16/2025]
Abstract
Purpose While prognostic models for retinal vein occlusion (RVO) exist, subtype-specific risk prediction tools for central retinal vein occlusion (CRVO) and branch retinal vein occlusion (BRVO) remain limited. This study aimed to construct and validate distinct CRVO and BRVO risk stratification nomograms. Methods We retrospectively analyzed electronic medical records from a tertiary hospital in Guangzhou (January 2010-November 2024). Non-RVO controls were matched 1:4 (CRVO) and 1:2 (BRVO) by sex and year of admission. The final cohorts included 630 patients (126 CRVO cases and 504 controls) and 813 patients (271 BRVO cases and 542 controls). Predictors encompassed clinical histories and laboratory indices. Multivariate regression identified independent risk factors, and model performance was evaluated using the area under the receiver operating characteristic curve (AUC), calibration plots, and decision curve analysis (DCA). Results The CRVO-nom and BRVO-nom highlighted significant predictors, including the neutrophil-to-lymphocyte ratio (NLR). Additional risk factors for CRVO included high-density lipoprotein cholesterol (HDL-C), platelet distribution width (PDW), history of diabetes, cerebral infarction, and coronary artery disease (CAD). For BRVO, significant predictors included a history of hypertension, age, and body mass index (BMI). The AUC for CRVO-nom was 0.80 (95% CI: 0.73-0.87) in the training set and 0.77 (95% CI: 0.65-0.86) in the validation set, while BRVO-nom yielded an AUC of 0.95 (95 %CI: 0.91-0.97) in the training set and 0.95 (95% CI: 0.89-0.98) in the validation set. Conclusions CRVO and BRVO exhibit distinct risk profiles. The developed nomograms-CRVO-nom and BRVO-nom-provide subtype-specific risk stratification with robust discrimination and clinical applicability. An online Shiny calculator facilitates real-time risk estimation, enabling targeted prevention for high-risk populations.
Collapse
Affiliation(s)
- Chunlan Liang
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Lian Liu
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Wenjuan Yu
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Qi Shi
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Jiang Zheng
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Jun Lyu
- Department of Clinical Research, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Jingxiang Zhong
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Department of Ophthalmology, The Sixth Affiliated Hospital of Jinan University, Dongguan, China
| |
Collapse
|
7
|
Zhang J, Miao C, Zhang H. Targeting neutrophil extracellular traps in cancer progression and metastasis. Theranostics 2025; 15:5846-5869. [PMID: 40365275 PMCID: PMC12068306 DOI: 10.7150/thno.111096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Accepted: 04/07/2025] [Indexed: 05/15/2025] Open
Abstract
Neutrophils serve as pivotal effectors and regulators of the intricate immune system. Their contributions are indispensable, encompassing the obliteration of pathogens and a significant role in both cancer initiation and progression. Conversely, malignancies profoundly affect neutrophil activity, maturation, and lifespans. Cancer cells manipulate their biology to enhance or suppress the key functions of neutrophils. This manipulation is one of the most remarkable defensive mechanisms used by neutrophils, including the formation of neutrophil extracellular traps (NETs). NETs are filamentous structures comprising DNA, histones, and proteins derived from cytotoxic granules. In this review, we discuss the bidirectional interplay in which cancer elicits NET formation, and NETs concurrently facilitate cancer progression. Here, we discuss how vascular dysfunction and thrombosis induced by neutrophils and NETs contribute to an elevated risk of mortality from cardiovascular complications in patients with cancer. Ultimately, we propose a series of therapeutic strategies that hold promise for effectively targeting NETs in clinical settings.
Collapse
Affiliation(s)
- Ji Zhang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
- Department of Anesthesiology, Shanghai Medical College, Fudan University, China
| | - Changhong Miao
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
- Department of Anesthesiology, Shanghai Medical College, Fudan University, China
| | - Hao Zhang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
- Department of Anesthesiology, Shanghai Medical College, Fudan University, China
| |
Collapse
|
8
|
Ma Y, Zhang Z, Cao X, Guo D, Huang S, Xie L, Wu M, Li J, Li C, Chu Y, Jiang S, Hao Y, Wang C, Zhong X, Ju R, Zhang F, Liu C, Wei Y. Semaphorin 6A phase separation sustains a histone lactylation-dependent lactate buildup in pathological angiogenesis. Proc Natl Acad Sci U S A 2025; 122:e2423677122. [PMID: 40244673 PMCID: PMC12036978 DOI: 10.1073/pnas.2423677122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 02/25/2025] [Indexed: 04/18/2025] Open
Abstract
Ischemic retinal diseases are major causes of blindness worldwide and are characterized by pathological angiogenesis. Epigenetic alterations in response to metabolic shifts in endothelial cells (ECs) suffice to underlie excessive angiogenesis. Lactate accumulation and its subsequent histone lactylation in ECs contribute to vascular disorders. However, the regulatory mechanism of establishing and sustaining lactylation modification remains elusive. Here, we showed that lactate accumulation induced histone lactylations on H3K9 and H3K18 in neovascular ECs in the proliferative stage of oxygen-induced retinopathy. Joint CUT&Tag and scRNA-seq analyses identified Prmt5 as a target of H3K9la and H3K18la in isolated retinal ECs. EC-specific deletion of Prmt5 since the early stage of revascularization suppressed a positive feedback loop of lactate production and histone lactylation, thus inhibiting neovascular tuft formation. Mechanistically, the C-terminal intrinsically disorder region (IDR) of the transmembrane semaphorin 6A (SEMA6A) forms liquid-liquid phase separation condensates to recruit RHOA and P300, facilitating P300 phosphorylation and histone lactylation cycle. Deletion of endothelial Sema6A reduced H3K9la and H3K18la at the promoter of PRMT5 and diminished its expression. The induction of histone lactylation by SEMA6A-IDR and its pro-angiogenic effect were abrogated by deletion of Prmt5. Our study illustrates a sustainable histone lactylation machinery driven by phase separation-dependent lactyltransferase activation in dysregulated vascularization.
Collapse
Affiliation(s)
- Ya Ma
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou510080, China
| | - Zhuyi Zhang
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou510080, China
| | - Xiaolian Cao
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou510080, China
| | - Dianlei Guo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou510060, China
| | - Shuting Huang
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou510080, China
| | - Lijing Xie
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou510060, China
| | - Mingjuan Wu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou510060, China
| | - Junru Li
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou510080, China
| | - Chenxin Li
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou510080, China
| | - Yu Chu
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou510080, China
| | - Shuxin Jiang
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou510080, China
| | - Yu Hao
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou510080, China
| | - Can Wang
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou510080, China
| | - Xiali Zhong
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou510080, China
| | - Rong Ju
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou510060, China
| | - Feng Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou510060, China
| | - Chunqiao Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou510060, China
| | - Yanhong Wei
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou510080, China
| |
Collapse
|
9
|
Lu Y, Zhang Y, Yao J, Bai W, Li K. Histone Modifications: Potential Therapeutic Targets for Diabetic Retinopathy. Biomolecules 2025; 15:575. [PMID: 40305347 PMCID: PMC12024956 DOI: 10.3390/biom15040575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Revised: 04/01/2025] [Accepted: 04/10/2025] [Indexed: 05/02/2025] Open
Abstract
Diabetic retinopathy (DR) is a microvascular complication arising as a secondary effect of diabetes, with both genetic and environmental factors playing a significant role in its onset and progression. Epigenetics serves as the crucial link between these genetic and environmental influences. Among the various epigenetic mechanisms, histone modification stands out as a key regulatory process associated with the development of many diseases. Histone modifications primarily regulate cellular function by influencing gene expression. Modulating histone modifications, particularly through the regulation of enzymes involved in these processes, holds a promising therapeutic approach for managing diseases like DR. In this review, we explore the regulatory mechanisms of histone modification and its contribution to the pathogenesis of DR.
Collapse
Affiliation(s)
- Yao Lu
- Department of Ophthalmology, The Affiliated Eye Hospital of Nanjing Medical University, Nanjing 210029, China; (Y.L.); (J.Y.)
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing 210029, China;
| | - Yizheng Zhang
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing 210029, China;
| | - Jin Yao
- Department of Ophthalmology, The Affiliated Eye Hospital of Nanjing Medical University, Nanjing 210029, China; (Y.L.); (J.Y.)
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing 210029, China;
| | - Wen Bai
- Department of Ophthalmology, The Affiliated Eye Hospital of Nanjing Medical University, Nanjing 210029, China; (Y.L.); (J.Y.)
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing 210029, China;
| | - Keran Li
- Department of Ophthalmology, The Affiliated Eye Hospital of Nanjing Medical University, Nanjing 210029, China; (Y.L.); (J.Y.)
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing 210029, China;
| |
Collapse
|
10
|
Chen X, Rong Y, Jiang Y, Zhang Q, Xiang S, Chen Z, Chen W, Zhang H, Deng C, Wang J. Vitamin K1 Alleviates Retinal Inflammation Following Acute Ocular Hypertension by Modulating Microglial Ferroptosis. Invest Ophthalmol Vis Sci 2025; 66:46. [PMID: 40244608 PMCID: PMC12013678 DOI: 10.1167/iovs.66.4.46] [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: 10/13/2024] [Accepted: 03/13/2025] [Indexed: 04/18/2025] Open
Abstract
Purpose Glaucoma is the leading cause of irreversible blindness worldwide and encompasses a group of diseases characterized by optic nerve atrophy and visual field defects. Acute intraocular pressure (IOP) elevation is a key driver of retinal inflammation and optic nerve damage, often accompanied by microglial activation and dysregulated ferroptosis pathways. Vitamin K1, a fat-soluble vitamin, possesses anti-inflammatory and antioxidant properties, and has the potential to regulate ferroptosis. However, its mechanisms in alleviating retinal inflammation following acute IOP elevation remain unclear. Methods In vivo, we established a mouse model of acute ocular hypertension to evaluate the protective effects of vitamin K1 on the retina and visual function. Transcriptome sequencing was used to explore the underlying mechanisms by which vitamin K1 exerts its effects. Immunofluorescence and Western blot were used to assess retinal inflammation and observe ferroptosis in microglia. In vitro, we developed a BV2 cell OGDR model to investigate the regulatory effects of vitamin K1 on iron metabolism and inflammation in microglia. Results Our findings demonstrated that acute IOP elevation led to microglial activation, along with iron overload and ferroptosis in microglia. Further analyses revealed that microglial ferroptosis was accompanied by an upregulation of inflammatory cytokine gene expression and protein levels. Vitamin K1 intervention, however, inhibited microglial ferroptosis, alleviated retinal inflammation, minimized retinal ganglion cell (RGC) loss, and protected visual function. Conclusions In conclusion, this study demonstrates that vitamin K1 exerts a protective effect by modulating microglial ferroptosis, thereby alleviating acute ocular hypertension-induced retinal inflammation.
Collapse
Affiliation(s)
- Xi Chen
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yan Rong
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yuxian Jiang
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qiuxiang Zhang
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Sifei Xiang
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhiqi Chen
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wei Chen
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hong Zhang
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Chaohua Deng
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Junming Wang
- Ophthalmic Center, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
11
|
Floyd JL, Prasad R, Dupont MD, Adu-Rutledge Y, Anshumali S, Paul S, Li Calzi S, Qi X, Malepati A, Johnson E, Jumbo-Lucioni P, Crosson JN, Mason JO, Boulton ME, Welner RS, Grant MB. Intestinal neutrophil extracellular traps promote gut barrier damage exacerbating endotoxaemia, systemic inflammation and progression of diabetic retinopathy in type 2 diabetes. Diabetologia 2025; 68:866-889. [PMID: 39875729 PMCID: PMC11950064 DOI: 10.1007/s00125-024-06349-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 11/01/2024] [Indexed: 01/30/2025]
Abstract
AIMS/HYPOTHESIS Within the small intestine, neutrophils play an integral role in preventing bacterial infection. Upon interaction with bacteria or bacteria-derived antigens, neutrophils initiate a multi-staged response of which the terminal stage is NETosis, formation of protease-decorated nuclear DNA into extracellular traps. NETosis has a great propensity to elicit ocular damage and has been associated with diabetic retinopathy and diabetic macular oedema (DME) progression. Here, we interrogate the relationship between gut barrier dysfunction, endotoxaemia and systemic and intestinal neutrophilia in diabetic retinopathy. METHODS In a cohort of individuals with type 2 diabetes (n=58) with varying severity of diabetic retinopathy and DME, we characterised the abundance of circulating neutrophils by flow cytometry and markers of gut permeability and endotoxaemia by plasma ELISA. In a mouse model of type 2 diabetes, we examined the effects of diabetes on abundance and function of intestinal, blood and bone marrow neutrophils, gut barrier integrity, endotoxaemia and diabetic retinopathy severity. Pharmacological inhibition of NETosis was achieved by i.p. injection of the peptidyl arginine deiminase 4 inhibitor (PAD4i) GSK484 daily for 4 weeks between 6 and 7 months of type 2 diabetes. RESULTS In human participants, neutrophilia was unique to individuals with type 2 diabetes with diabetic retinopathy and DME and was accompanied by heightened circulating markers of gut permeability. At late-stage diabetes, neutrophilia and gut barrier dysfunction were seen in db/db mice. The db/db mice exhibited an increase in stem-like pre-neutrophils in the intestine and bone marrow and a decrease in haematopoietic vascular reparative cells. In the db/db mouse intestine, enhanced loss of gut barrier integrity was associated with elevated intestinal NETosis. Inhibition of NETosis by the PAD4i GSK484 resulted in decreased abundance of premature neutrophils in the intestine and blood and resulted in neutrophil retention in the bone marrow compared with vehicle-treated db/db mice. Additionally, the PAD4i decreased senescence within the gut epithelium and yielded a slowing of diabetic retinopathy progression. CONCLUSIONS/INTERPRETATION Severity of diabetic retinopathy and DME were associated with peripheral neutrophilia, gut barrier dysfunction and endotoxaemia in the human participants. db/db mice exhibited intestinal neutrophilia, specifically stem-like pre-neutrophils, which was associated with elevated NETosis and decreased levels of vascular reparative cells. Chronic inhibition of NETosis in db/db mice reduced intestinal senescence and NETs in the retina. These changes were associated with reduced endotoxaemia and an anti-inflammatory bone marrow milieu with retention of pre-neutrophils in the bone marrow and increased gut infiltration of myeloid angiogenic cells. Collectively, PAD-4i treatment decreased gut barrier dysfunction, restoring physiological haematopoiesis and levels of haematopoietic vascular reparative cells.
Collapse
Affiliation(s)
- Jason L Floyd
- Department of Ophthalmology and Visual Sciences, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ram Prasad
- Department of Ophthalmology and Visual Sciences, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Mariana D Dupont
- Department of Ophthalmology and Visual Sciences, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Yvonne Adu-Rutledge
- Department of Ophthalmology and Visual Sciences, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Shambhavi Anshumali
- Department of Ophthalmology and Visual Sciences, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Sarbodeep Paul
- Department of Ophthalmology and Visual Sciences, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Sergio Li Calzi
- Department of Ophthalmology and Visual Sciences, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Xiaoping Qi
- Department of Ophthalmology and Visual Sciences, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Akanksha Malepati
- Department of Ophthalmology and Visual Sciences, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Emory Johnson
- Department of Ophthalmology and Visual Sciences, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Patricia Jumbo-Lucioni
- Pharmaceutical, Social and Administrative Sciences, Samford University, Birmingham, AL, USA
| | - Jason N Crosson
- Department of Ophthalmology and Visual Sciences, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Retina Consultants of Alabama, Birmingham, AL, USA
| | - John O Mason
- Department of Ophthalmology and Visual Sciences, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Retina Consultants of Alabama, Birmingham, AL, USA
| | - Michael E Boulton
- Department of Ophthalmology and Visual Sciences, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Robert S Welner
- Department of Medicine, Division Hematology/Oncology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Maria B Grant
- Department of Ophthalmology and Visual Sciences, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
12
|
Chen Y, Jiang F, Zeng Y, Zhang M. The role of retinal pigment epithelial senescence and the potential of senotherapeutics in age-related macular degeneration. Surv Ophthalmol 2025:S0039-6257(25)00053-0. [PMID: 40089029 DOI: 10.1016/j.survophthal.2025.03.004] [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: 10/14/2024] [Revised: 03/03/2025] [Accepted: 03/10/2025] [Indexed: 03/17/2025]
Abstract
Age-related macular degeneration (AMD) is a leading cause of visual impairment in the aging population. Evidence showing the presence of cellular senescence in retinal pigment epithelium (RPE) of patients with AMD is growing. Senescent RPE play a pivotal role in its pathogenesis. The senescent RPE suffers from structural and functional alterations and disruption of the surrounding microenvironment due to the development of the senescence-associated secretory phenotype, which contributes to metabolic dysfunctions and inflammatory responses in the retina. Senotherapeutics, including senolytics, senomorphics and others, are novel treatments targeting senescent cells and are promising treatments for AMD. As senotherapeutic targets are being developed, it is promising that the burden of AMD could be decreased.
Collapse
Affiliation(s)
- Yingying Chen
- Department of Ophthalmology and Laboratory of Macular Disease, West China Hospital, Sichuan University, China.
| | - Feipeng Jiang
- Department of Ophthalmology and Laboratory of Macular Disease, West China Hospital, Sichuan University, China.
| | - Yue Zeng
- Department of Ophthalmology and Laboratory of Macular Disease, West China Hospital, Sichuan University, China.
| | - Meixia Zhang
- Department of Ophthalmology and Laboratory of Macular Disease, West China Hospital, Sichuan University, China.
| |
Collapse
|
13
|
Liu D, Meng Z, Jin C, Chen F, Pu L, Wu Z, Zeng Q, Luo J, Wu W. Fibronectin Mediates Endothelial-to-Mesenchymal Transition in Retina Angiogenesis. Invest Ophthalmol Vis Sci 2025; 66:10. [PMID: 40042877 PMCID: PMC11892531 DOI: 10.1167/iovs.66.3.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 02/09/2025] [Indexed: 03/12/2025] Open
Abstract
Purpose The purpose of this study was to investigate the role of endothelial-mesenchymal transition (EndoMT) in pathological retinal angiogenesis and identify key molecular mediators in retina angiogenesis. Methods RNA sequencing (RNA-seq) was performed on retinal tissue from an oxygen-induced retinopathy (OIR) mouse model to analyze gene expression patterns. The Gene Set Enrichment Analysis was used to examine the correlation between epithelial-mesenchymal transition (EMT) and angiogenesis gene sets. Fibronectin (FN1) expression was evaluated in endothelial cells, and its function was assessed through siRNA-mediated knockdown in both in vitro angiogenesis assays and the OIR model. Results EndoMT occurred early in retinal angiogenesis development, with significant correlation between EMT and angiogenesis gene sets. FN1 was identified as the most significantly upregulated EMT-related gene in endothelial cells. The siRNA-mediated inhibition of fibronectin effectively prevented VEGF-induced angiogenesis in vitro and reduced pathological angiogenesis in the OIR model. Conclusions EndoMT is a crucial early event in pathological retinal angiogenesis, with fibronectin serving as a key mediator. Targeting fibronectin may provide a novel therapeutic strategy that could synergize with anti-VEGF treatments to more effectively treat pathological angiogenesis in diabetic retinopathy (DR) and retinopathy of prematurity (ROP), particularly in cases of poor response to anti-VEGF therapy alone.
Collapse
Affiliation(s)
- Dan Liu
- Department of Ophthalmology, Hunan Key Laboratory of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Zhishang Meng
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Chen Jin
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Fang Chen
- Huan Key Laboratory of Molecular Precision Medicine, Xiangya Hospital and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Li Pu
- Department of Ophthalmology, Hunan Key Laboratory of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China
| | - Ze Wu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Qi Zeng
- Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Hunan, China
| | - Jing Luo
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Wenyi Wu
- Department of Ophthalmology, Hunan Key Laboratory of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China
| |
Collapse
|
14
|
Nian S, Zeng Y, Heyden KE, Cagnone G, Yagi H, Boeck M, Lee D, Hirst V, Hua Z, Lee J, Wang C, Neilsen K, Joyal JS, Field MS, Fu Z. Folic Acid Supplementation Inhibits Proliferative Retinopathy of Prematurity. Biomolecules 2025; 15:309. [PMID: 40001612 PMCID: PMC11852370 DOI: 10.3390/biom15020309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 02/11/2025] [Accepted: 02/17/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Retinopathy of prematurity (ROP) is the major cause of blindness in children. It is a biphasic disease with retinal vessel growth cessation and loss (Phase I) followed by uncontrolled retinal vessel growth (Phase II). Folate is an essential nutrient for fetal development and growth. Premature infants have a high risk for folate deficiency. However, the contribution of folate to ROP risk remains unknown. METHODS In mouse oxygen-induced retinopathy (OIR), the nursing dams were fed with a folic acid-deficient or control diet after delivery until the end of hyperoxia. Alternatively, pups received direct injection of either folic acid or vehicle during Phase I hyperoxia. Genes involved in the folate cycle and angiogenic responses were examined using real-time PCR. Total retinal folate levels were measured with the Lactobacillus casei assay. RESULTS Maternal folic acid deficiency in early life exacerbated pathological retinal vessel growth, while supplementation with folic acid suppressed it. Genes involved in the folate cycle were downregulated in Phase I OIR retinas and were highly expressed in Müller glia. Folic acid reduced pro-angiogenic signaling in cultured rat retinal Müller glia in vitro. CONCLUSIONS Appropriate supplementation of folic acid might be a new and safe treatment for ROP at an early stage.
Collapse
Affiliation(s)
- Shen Nian
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (S.N.); (Y.Z.); (H.Y.); (D.L.); (V.H.); (Z.H.); (C.W.); (K.N.)
- Department of Pathology, Xi’an Medical University, Xi’an 710021, China
| | - Yan Zeng
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (S.N.); (Y.Z.); (H.Y.); (D.L.); (V.H.); (Z.H.); (C.W.); (K.N.)
| | - Katarina E. Heyden
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA; (K.E.H.); (M.S.F.)
| | - Gaël Cagnone
- Centre Hospitalier Universitaire Sainte-Justine Research Center, Montreal, QC H3T 1C5, Canada (J.-S.J.)
| | - Hitomi Yagi
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (S.N.); (Y.Z.); (H.Y.); (D.L.); (V.H.); (Z.H.); (C.W.); (K.N.)
- Department of Ophthalmology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Myriam Boeck
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (S.N.); (Y.Z.); (H.Y.); (D.L.); (V.H.); (Z.H.); (C.W.); (K.N.)
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Deokho Lee
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (S.N.); (Y.Z.); (H.Y.); (D.L.); (V.H.); (Z.H.); (C.W.); (K.N.)
| | - Victoria Hirst
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (S.N.); (Y.Z.); (H.Y.); (D.L.); (V.H.); (Z.H.); (C.W.); (K.N.)
| | - Zhanqing Hua
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (S.N.); (Y.Z.); (H.Y.); (D.L.); (V.H.); (Z.H.); (C.W.); (K.N.)
| | - Jeff Lee
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (S.N.); (Y.Z.); (H.Y.); (D.L.); (V.H.); (Z.H.); (C.W.); (K.N.)
| | - Chaomei Wang
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (S.N.); (Y.Z.); (H.Y.); (D.L.); (V.H.); (Z.H.); (C.W.); (K.N.)
| | - Katherine Neilsen
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (S.N.); (Y.Z.); (H.Y.); (D.L.); (V.H.); (Z.H.); (C.W.); (K.N.)
| | - Jean-Sébastien Joyal
- Centre Hospitalier Universitaire Sainte-Justine Research Center, Montreal, QC H3T 1C5, Canada (J.-S.J.)
- Department of Pediatrics, Faculty of Medicine, Université de Montréal, Montreal, QC H3C 3J7, Canada
| | - Martha S. Field
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA; (K.E.H.); (M.S.F.)
| | - Zhongjie Fu
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (S.N.); (Y.Z.); (H.Y.); (D.L.); (V.H.); (Z.H.); (C.W.); (K.N.)
| |
Collapse
|
15
|
Song H, Li Q, Gui X, Fang Z, Zhou W, Wang M, Jiang Y, Geng A, Shen X, Liu Y, Zhang H, Nie Z, Zhang L, Zhu H, Zhang F, Li X, Luo F, Zhang H, Shen W, Sun X. Endothelial protein C receptor promotes retinal neovascularization through heme catabolism. Nat Commun 2025; 16:1603. [PMID: 39948347 PMCID: PMC11825934 DOI: 10.1038/s41467-025-56810-0] [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: 03/29/2024] [Accepted: 02/03/2025] [Indexed: 02/16/2025] Open
Abstract
Pathological retinal neovascularization (RNV) is one of the leading causes of blindness worldwide; however, its underlying mechanism remains unclear. Here, we found that the expression of endothelial protein C receptor (Epcr) was increased during RNV, and its ligand was elevated in the serum or vitreous body of patients with proliferative diabetic retinopathy. Deleting endothelial Epcr or using an EPCR-neutralizing antibody ameliorated pathological retinal angiogenesis. EPCR promoted endothelial heme catabolism and carbon monoxide release through heme oxygenase 1 (HO-1). Inhibition of heme catabolism by deleting endothelial Ho-1 or using an HO-1 inhibitor suppressed pathological angiogenesis in retinopathy. Conversely, supplementation with carbon monoxide rescued the angiogenic defects after endothelial Epcr or Ho-1 deletion. Our results identified EPCR-dependent endothelial heme catabolism as an important contributor to pathological angiogenesis, which may serve as a potential target for treating vasoproliferative retinopathy.
Collapse
Affiliation(s)
- Hongyuan Song
- Department of Ophthalmology, Shanghai Changhai Hospital, Shanghai, China.
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- National Clinical Research Center for Ophthalmic Diseases, Shanghai, China.
- Oriental Pan-Vascular Devices Innovation College, University of Shanghai for Science and Technology, Shanghai, China.
| | - Qing Li
- Department of Ophthalmology, Shanghai Changhai Hospital, Shanghai, China
- The Affiliated Eye Hospital of Nanjing Medical University, Nanjing, China
| | - Xiao Gui
- Department of Ophthalmology, Shanghai Changhai Hospital, Shanghai, China
- Department of Ophthalmology, Yuanwang Hospital, Wuxi, China
| | - Ziyu Fang
- Department of Urology, Shanghai Changhai Hospital, Shanghai, China
| | - Wen Zhou
- Department of Ophthalmology, Shanghai Changhai Hospital, Shanghai, China
| | - Mengzhu Wang
- Department of Ophthalmology, Shanghai Changhai Hospital, Shanghai, China
| | - Yuxin Jiang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Fundus Diseases, Shanghai, China
| | - Ajun Geng
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, Hangzhou, China
- New Cornerstone Science Laboratory, State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Xi Shen
- Department of Ophthalmology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yongxuan Liu
- Department of Ophthalmology, Shanghai Changhai Hospital, Shanghai, China
| | - Haorui Zhang
- Department of Ophthalmology, Shanghai Changhai Hospital, Shanghai, China
| | - Zheng Nie
- Department of Ophthalmology, Shanghai Changhai Hospital, Shanghai, China
| | - Lin Zhang
- Department of Ophthalmology, Shanghai Changhai Hospital, Shanghai, China
| | - Huimin Zhu
- Department of Ophthalmology, Shanghai Changhai Hospital, Shanghai, China
| | - Feng Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Xuri Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Fanyan Luo
- Department of Cardiac Surgery, Xiangya Hospital, Central South University, Changsha, China.
| | - Hongjian Zhang
- Oriental Pan-Vascular Devices Innovation College, University of Shanghai for Science and Technology, Shanghai, China.
- Shidong Hospital Affiliated to University of Shanghai for Science and Technology, 999 Shiguang Road, Shanghai, China.
| | - Wei Shen
- Department of Ophthalmology, Shanghai Changhai Hospital, Shanghai, China.
| | - Xiaodong Sun
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- National Clinical Research Center for Ophthalmic Diseases, Shanghai, China.
- Shanghai Key Laboratory of Fundus Diseases, Shanghai, China.
| |
Collapse
|
16
|
Wang X, Wang J, Huang L, Huang G. Capsiate Improves Glucose Metabolism by Improving Insulin Sensitivity in Diabetic Retinopathy Mice. Curr Eye Res 2025; 50:213-220. [PMID: 39431723 DOI: 10.1080/02713683.2024.2412296] [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: 06/03/2024] [Revised: 09/23/2024] [Accepted: 09/30/2024] [Indexed: 10/22/2024]
Abstract
PURPOSE Capsiate (cap) is a metabolite that affects a number of biological processes, and diabetic retinopathy (DR) is now known to be the primary cause of end-stage eye illness. METHODS In order to examine the effects of the cap intervention on body weight, nutritional intake, changes in body weight composition, glucose metabolism levels, retinopathy, and oxidative stress levels, we proposed using a mouse model of diabetic retinopathy caused by STZ. RESULTS Our findings demonstrated that, in addition to increasing lean body mass and lowering fat body mass content, cap intervention significantly improved body weight and dietary consumption in STZ mice. Additionally, our results on glucose metabolism revealed that cap had a significant impact on insulin resistance and the stabilization of OGTT levels. In conclusion, we examined the levels of oxidative stress and retinopathy. We discovered that the cap intervention greatly reduced the levels of MDA and significantly improved the levels of VEGF and retinopathy. In contrast, the STZ group's levels of SOD, CAT, and GSH were significantly higher. CONCLUSIONS According to our research, the Cap intervention improved the damage caused by diabetic retinopathy by reversing the levels of oxidative stress and the disrupted state of glucose metabolism, which in turn decreased the levels of VEGF.
Collapse
Affiliation(s)
- Xiaorui Wang
- Department of Ophthalmology, Anhui Medical University Affiliated Lu'an People's Hospital, Lu'an City, Fujian Province, China
| | - Jingwen Wang
- Department of Nutrition, Quanzhou Medical College, Quanzhou City, Fujian Province, China
| | - Lijuan Huang
- Department of Ophthalmology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - Guangqian Huang
- Department of Ophthalmology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| |
Collapse
|
17
|
Xu Y, Chen L, Liu W, Chen L. [Advances in inflammaging in liver disease]. Zhejiang Da Xue Xue Bao Yi Xue Ban 2025; 54:90-98. [PMID: 39828280 PMCID: PMC11956859 DOI: 10.3724/zdxbyxb-2024-0249] [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: 06/04/2024] [Accepted: 11/25/2024] [Indexed: 01/22/2025]
Abstract
Inflammaging is a process of cellular dysfunction associated with chronic inflammation, which plays a significant role in the onset and progression of liver diseases. Research on its mechanisms has become a hotspot. In viral hepatitis, inflammaging primarily involve oxidative stress, cell apoptosis and necrosis, as well as gut microbiota dysbiosis. In non-alcoholic fatty liver disease, inflammaging is more complex, involving insulin resistance, fat deposition, lipid metabolism disorders, gut microbiota dysbiosis, and abnormalities in NAD+ metabolism. In liver tumors, inflammaging is characterized by weakening of tumor suppressive mechanisms, remodeling of the liver microenvironment, metabolic reprogramming, and enhanced immune evasion. Therapeutic strategies targeting inflammaging have been developing recently, and antioxidant therapy, metabolic disorder improvement, and immunotherapy are emerging as important interventions for liver diseases. This review focuses on the mechanisms of inflammaging in liver diseases, aiming to provide novel insights for the prevention and treatment of liver diseases.
Collapse
Affiliation(s)
- Yanping Xu
- Department of General Practice, Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China.
| | - Luyi Chen
- Department of General Practice, Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Weili Liu
- Department of General Practice, Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Liying Chen
- Department of General Practice, Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China.
| |
Collapse
|
18
|
Jarzebska N, Rodionov RN, Voit-Bak K, Straube R, Mücke A, Tselmin S, Rettig R, Julius U, Siow R, Gräßler J, Passauer J, Kok Y, Mavberg P, Weiss N, Bornstein SR, Aswani A. Neutrophil Extracellular Traps (NETs) as a Potential Target for Anti-Aging: Role of Therapeutic Apheresis. Horm Metab Res 2025. [PMID: 39788160 DOI: 10.1055/a-2444-3422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
Neutrophil extracellular traps (NETs) are large structures composed of chromatin, histones and granule-derived proteins released extracellularly by neutrophils. They are generally considered to be a part of the antimicrobial defense strategy, preventing the dissemination of pathogens. However, overproduction of NETs or their ineffective clearance can drive various pathologies, many of which are associated with advanced age and involve uncontrolled inflammation, oxidative, cardiovascular and neurodegenerative stress as underlying mechanisms. Targeting NETs in the elderly as an anti-aging therapy seems to be a very attractive therapeutic approach. Therapeutic apheresis with a specific filter to remove NETs could be a promising strategy worth considering.
Collapse
Affiliation(s)
- Natalia Jarzebska
- Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Roman N Rodionov
- Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
- Department of Clinical Pharmacology, College of Medicine and Public Health, Flinders University and Flinders Medical Centre, Adelaide, Australia
| | - Karin Voit-Bak
- Zentrum für Apherese- und Hämofiltration, INUS Tagesklinikum, Cham, Germany
| | - Richard Straube
- Zentrum für Apherese- und Hämofiltration, INUS Tagesklinikum, Cham, Germany
| | - Anna Mücke
- INUSpheresis Center Basel, Ayus Medical Group, Basel, Switzerland
| | - Sergey Tselmin
- Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Ronny Rettig
- Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Ulrich Julius
- Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Richard Siow
- School of Cardiovascular and Metabolic Medicine and Sciences, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom of Great Britain and Northern Ireland
- Ageing Research at King's (ARK), King's College London, London, United Kingdom of Great Britain and Northern Ireland
- Department of Physiology, Anatomy and Genetics, Medical Sciences Division, University of Oxford, Oxford, United Kingdom of Great Britain and Northern Ireland
| | - Jürgen Gräßler
- Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Jens Passauer
- Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | | | - Philip Mavberg
- INUSpheresis Center Basel, Ayus Medical Group, Basel, Switzerland
| | - Norbert Weiss
- Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Stefan R Bornstein
- Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
- School of Cardiovascular and Metabolic Medicine and Sciences, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom of Great Britain and Northern Ireland
| | - Andrew Aswani
- Department of Intensive Care Medicine, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom of Great Britain and Northern Ireland
- Santersus AG, Zurich, Switzerland
| |
Collapse
|
19
|
Bu S, Ling JY, Wu X, Zhang L, Shi X, Huang L, Zhao Z, Yang Y, Xiang Z, Liu YU, Liu Y, Zhang Y. Downregulation of MerTK in circulating T cells of patients with non-proliferative diabetic retinopathy. Front Endocrinol (Lausanne) 2025; 15:1509445. [PMID: 39845889 PMCID: PMC11750652 DOI: 10.3389/fendo.2024.1509445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 12/09/2024] [Indexed: 01/24/2025] Open
Abstract
Objective To explore the differential gene expression in peripheral blood immune cells of individuals with type 2 diabetes mellitus (DM), comparing those with and without non-proliferative diabetic retinopathy (NPDR). Methods From a pool of 126 potential participants, 60 were selected for detailed analysis. This group included 12 healthy donors (HDs), 22 individuals with DM, and 26 with NPDR. We analyzed peripheral blood mononuclear cells (PBMCs) using RNA sequencing and quantitative PCR (qPCR) to pinpoint differentially expressed genes (DEGs). Western blot and flow cytometry were also employed to evaluate the protein expression of specific genes. Results In patients with NPDR compared to those with DM alone, MerTK-a gene implicated in inherited retinal dystrophies due to its mutations-was notably downregulated in PBMCs. Through flow cytometry, we assessed the protein levels and cellular distribution of MerTK, finding a predominant expression in monocytes and myeloid-derived suppressor cells (MDSCs), with a marked reduction in CD4+ and CD8+ T cells, as well as in natural killer T (NKT) cells. Patients with DM demonstrated a significant deviation in the PBMCs composition, particularly in B cells, CD4+ T cells, and NK cells, when compared to HDs. Conclusions The study indicates that MerTK expression in T cells within PBMCs could act as a viable blood biomarker for NPDR risk in patients with DM. Furthermore, the regulation of T cells by MerTK might represent a critical pathway through which DM evolves into NPDR.
Collapse
Affiliation(s)
- Shimiao Bu
- Department of Ophthalmology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Laboratory for Neuroimmunology in Health and Diseases, Center for Medical Research on Innovation and Translation, Institute of Clinical Medicine, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Jiang-Yue Ling
- Department of Ophthalmology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Laboratory for Neuroimmunology in Health and Diseases, Center for Medical Research on Innovation and Translation, Institute of Clinical Medicine, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Xiaojun Wu
- Department of Ophthalmology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Laboratory for Neuroimmunology in Health and Diseases, Center for Medical Research on Innovation and Translation, Institute of Clinical Medicine, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Liting Zhang
- Department of Ophthalmology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Laboratory for Neuroimmunology in Health and Diseases, Center for Medical Research on Innovation and Translation, Institute of Clinical Medicine, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Xiangyu Shi
- Department of Ophthalmology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Lang Huang
- Laboratory for Neuroimmunology in Health and Diseases, Center for Medical Research on Innovation and Translation, Institute of Clinical Medicine, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Zheng Zhao
- Department of Neurology, The First Affiliated Hospital, School of Medicine, Jinan University, Guangzhou, China
| | - Ying Yang
- Department of Neurology, The First Affiliated Hospital, School of Medicine, Jinan University, Guangzhou, China
| | - Zongqin Xiang
- Laboratory for Neuroimmunology in Health and Diseases, Center for Medical Research on Innovation and Translation, Institute of Clinical Medicine, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yong U. Liu
- Laboratory for Neuroimmunology in Health and Diseases, Center for Medical Research on Innovation and Translation, Institute of Clinical Medicine, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Department of Neurology, Multi-Omics Research Center for Brain Disorders, The First Affiliated Hospital, University of South China, Hengyang, China
| | - Yufeng Liu
- Laboratory for Neuroimmunology in Health and Diseases, Center for Medical Research on Innovation and Translation, Institute of Clinical Medicine, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yuehong Zhang
- Department of Ophthalmology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Laboratory for Neuroimmunology in Health and Diseases, Center for Medical Research on Innovation and Translation, Institute of Clinical Medicine, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| |
Collapse
|
20
|
Liu Q, Xia LX, Yi WZ, Wu YN, Gu SS, Chen JY, Liu TT, Lu YH, Cui YH, Meng J, Pan HW. Inhibition of Retinal Neovascularization by BEZ235: Targeting the Akt/4EBP1/Cyclin D1 Pathway in Endothelial Cells. Invest Ophthalmol Vis Sci 2025; 66:66. [PMID: 39888634 PMCID: PMC11784786 DOI: 10.1167/iovs.66.1.66] [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: 05/13/2024] [Accepted: 12/17/2024] [Indexed: 02/01/2025] Open
Abstract
Purpose To investigate the therapeutic efficacy of BEZ235, a dual PI3K/mTOR inhibitor, in suppressing pathological neovascularization in an oxygen-induced retinopathy (OIR) mouse model and explore the role of cyclin D1 in endothelial cell cycle regulation. Methods Single-cell RNA sequencing was performed to analyze gene expression and cell-cycle alterations in retinal endothelial cells under normoxic and OIR conditions. The effects of BEZ235 on human umbilical vein endothelial cells (HUVECs) and human retinal microvascular endothelial cells (HRMECs) were evaluated by assessing cell viability, cell-cycle progression, proliferation, migration, and tube formation. In the OIR mouse model, retinal neovascularization was evaluated by retinal flatmount immunofluorescence staining, hematoxylin and eosin (H&E) staining, quantitative reverse-transcription polymerase chain reaction (RT-qPCR), and western blot analyses. The in vivo toxicity of BEZ235 was evaluated by electroretinography (ERG) and histological examination of the heart, liver, spleen, lungs, and kidneys. Results In vitro, BEZ235 significantly inhibited cell cycle progression by downregulating cyclin D1 at both mRNA and protein levels, inducing G0/G1 phase arrest. This led to significant reductions in cell viability, proliferation, migration, and tube formation. In the OIR model, BEZ235 substantially decreased neovascularization and improved vascular organization. BEZ235 mediates its effects by inhibiting the PI3K/Akt/mTOR pathway, reducing Akt and 4E-binding protein 1 (4EBP1) phosphorylation levels, thus downregulating cyclin D1 expression. ERG and histological examination suggested that BEZ235 did not induce evident retinal or systemic toxicity at the dosage used to inhibit retinal neovascularization. Conclusions BEZ235 effectively inhibits retinal neovascularization by downregulating cyclin D1 via 4EBP1 phosphorylation inhibition, highlighting its potential as a promising therapeutic agent for retinal neovascularization diseases.
Collapse
Affiliation(s)
- Qi Liu
- Institute of Ophthalmology, School of Medicine, Jinan University, Guangzhou, China
- Department of Ophthalmology, the First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Ling-Xiao Xia
- Institute of Ophthalmology, School of Medicine, Jinan University, Guangzhou, China
- Department of Ophthalmology, the First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Wan-Zhao Yi
- Institute of Ophthalmology, School of Medicine, Jinan University, Guangzhou, China
- Department of Ophthalmology, the First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Ya-Ni Wu
- Institute of Ophthalmology, School of Medicine, Jinan University, Guangzhou, China
- Department of Ophthalmology, the First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Shuo-Shuo Gu
- Institute of Ophthalmology, School of Medicine, Jinan University, Guangzhou, China
- Department of Ophthalmology, the First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Jian-Ying Chen
- Institute of Ophthalmology, School of Medicine, Jinan University, Guangzhou, China
- Department of Ophthalmology, the First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Ting-Ting Liu
- Institute of Ophthalmology, School of Medicine, Jinan University, Guangzhou, China
- Department of Ophthalmology, the First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Ying-Hui Lu
- Institute of Ophthalmology, School of Medicine, Jinan University, Guangzhou, China
- Department of Ophthalmology, the First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Yu-Hong Cui
- Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Department of Histology and Embryology, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Jing Meng
- Department of Ophthalmology, the First Affiliated Hospital, Jinan University, Guangzhou, China
- The Affiliated Shunde Hospital of Jinan University, Foshan, China
| | - Hong-Wei Pan
- Institute of Ophthalmology, School of Medicine, Jinan University, Guangzhou, China
- Department of Ophthalmology, the First Affiliated Hospital, Jinan University, Guangzhou, China
| |
Collapse
|
21
|
Liang A, Feng T, Gao X, Zhao B, Chen S. Identification of PDGFA as a Neutrophil-related Biomarker Linked to the Advancement of Diabetic Retinopathy through Integrated Bioinformatics Analysis. Endocr Metab Immune Disord Drug Targets 2025; 25:109-121. [PMID: 38504565 DOI: 10.2174/0118715303279463240220050158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 01/18/2024] [Accepted: 01/30/2024] [Indexed: 03/21/2024]
Abstract
BACKGROUND The dysregulation of the innate immune system plays a crucial role in the development of Diabetic Retinopathy (DR). To gain an insight into the underlying mechanism of DR, it is essential to identify specific biomarkers associated with immune cell infiltration. METHODS In this study, we retrieved the GSE94019 and GSE60436 datasets from the Gene Expression Omnibus (GEO) database. By utilizing CIBERSORT, MCPcounter, and xCell algorithms, we conducted a comprehensive analysis of the immune cell infiltration landscape in DR. The limma package was employed to identify Differentially Expressed Necroptosis-related Genes (DENRGs). Subsequently, enrichment analysis was performed to investigate the potential functions of the DENRGs. To identify the core DENRGs, the CytoHubba plug-in in Cytoscape software was utilized. The expression levels of these core DENRGs were verified in an independent dataset. RESULTS Our analysis identified 213 DENRGs, and among them, Platelet-derived Growth Factor subunit A (PDGFA) was identified as a core DENRG. Notably, the expression of PDGFA was found to be upregulated in DR, and this finding was further validated in the GSE102485 dataset. Additionally, the results of GSVA and GSEA revealed that in the high PDGFA group, there was activation of pathways related to inflammation and the immune system. Moreover, analysis of immune infiltration demonstrated a significant association between PDGFA gene expression and the infiltration levels of specific immune cells, including basophils, macrophages M1, macrophages, neutrophils, monocytes, NK cells, and B cells. CONCLUSION The involvement of neutrophils in the development and progression of DR is suggested. PDGFA has emerged as a potential marker and is linked to the infiltration of immune cells in DR. These findings shed new light on the underlying mechanisms of DR.
Collapse
Affiliation(s)
- Anran Liang
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China
- Department of Ophthalmology, Jining No.1 People's Hospital, Jining, Shandong, China
| | - Tingting Feng
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin Eye Hospital, Tianjin, China
| | - Xiang Gao
- Anyang Eye Hospital, Anyang, Henan, China
| | - Bowen Zhao
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin Eye Hospital, Tianjin, China
| | - Song Chen
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin Eye Hospital, Tianjin, China
| |
Collapse
|
22
|
Zeng F, Shao Y, Wu J, Luo J, Yue Y, Shen Y, Wang Y, Shi Y, Wu D, Cata JP, Yang S, Zhang H, Miao C. Tumor metastasis and recurrence: The role of perioperative NETosis. Cancer Lett 2024; 611:217413. [PMID: 39725150 DOI: 10.1016/j.canlet.2024.217413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 12/11/2024] [Accepted: 12/22/2024] [Indexed: 12/28/2024]
Abstract
Although surgical resection of tumor mass remains the mainstay of curative therapeutic management for solid tumors, accumulating studies suggest that these procedures promote tumor recurrence and metastasis. Regarded as the first immune cells to fight against infectious or inflammatory insults from surgery, neutrophils along with their ability of neutrophil extracellular traps (NETs) production has attracted much attention. A growing body of evidence suggests that NETs promote cancer metastasis by stimulating various stages, including local invasion, colonization, and growth. Therefore, we discussed the mechanism of NETosis induced by surgical stress and tumor cells, and the contribution of NETs on tumor metastasis: aid in the tumor cell migration and proliferation, evasion of immune surveillance, circulating tumor cell adhesion and establishment of a metastatic niche. Lastly, we summarized existing NET-targeting interventions, offering recent insights into potential targets for clinical intervention.
Collapse
Affiliation(s)
- Fu Zeng
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Yuwen Shao
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Jingyi Wu
- Department of Anesthesiology, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China
| | - Jingwen Luo
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Ying Yue
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Yang Shen
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Yanghanzhao Wang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Yuxin Shi
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Dan Wu
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Juan P Cata
- Department of Anesthesiology and Perioperative Medicine, The University of Texas-MD Anderson Cancer Center, Houston, TX, USA; Anesthesiology and Surgical Oncology Research Group, Houston, TX, USA
| | - Shuofei Yang
- Department of Vascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Pujian Road 160, Shanghai, 200127, China.
| | - Hao Zhang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China.
| | - Changhong Miao
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China.
| |
Collapse
|
23
|
Hata M, Hata M, Dejda A, Pilon F, Diaz-Marin R, Fournier F, Joyal JS, Cagnone G, Ochi Y, Crespo-Garcia S, Wilson AM, Sapieha P. Corticosteroids reduce pathological angiogenesis yet compromise reparative vascular remodeling in a model of retinopathy. Proc Natl Acad Sci U S A 2024; 121:e2411640121. [PMID: 39693344 DOI: 10.1073/pnas.2411640121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 10/11/2024] [Indexed: 12/20/2024] Open
Abstract
Tissue inflammation is often broadly associated with cellular damage, yet sterile inflammation also plays critical roles in beneficial tissue remodeling. In the central nervous system, this is observed through a predominantly innate immune response in retinal vascular diseases such as age-related macular degeneration, diabetic retinopathy, and retinopathy of prematurity. Here, we set out to elucidate the dynamics of the immune response during progression and regression of pathological neovascularization in retinopathy. In a mouse model of oxygen-induced retinopathy, we report that dexamethasone, a broad-spectrum corticosteroid, suppresses initial formation of pathological preretinal neovascularization in early stages of disease, yet blunts reparative inflammation by impairing distinct myeloid cell populations, and hence reduces beneficial vascular remodeling in later stages of disease. Using genetic depletion of distinct components of the innate immune response, we demonstrate that CX3C chemokine receptor 1-expressing microglia contribute to angiogenesis. Conversely, myeloid cells expressing lysozyme 2 are recruited to sites of damaged blood vessels and pathological neovascularization where they partake in a reparative process that ultimately restores circulatory homeostasis to the retina. Hence, the Janus-faced properties of anti-inflammatory drugs should be considered, particularly in stages associated with persistent neovascularization.
Collapse
Affiliation(s)
- Masayuki Hata
- Departments of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, QC H1T 2M4, Canada
- Department of Biochemistry and Molecular Medicine, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, QC H1T 2M4, Canada
| | - Maki Hata
- Departments of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, QC H1T 2M4, Canada
| | - Agnieszka Dejda
- Departments of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, QC H1T 2M4, Canada
| | - Frédérique Pilon
- Departments of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, QC H1T 2M4, Canada
| | - Roberto Diaz-Marin
- Department of Biochemistry and Molecular Medicine, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, QC H1T 2M4, Canada
| | - Frédérik Fournier
- Departments of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, QC H1T 2M4, Canada
| | - Jean-Sebastien Joyal
- Departments of Pediatrics, Ophthalmology, and Pharmacology, Centre Hospitalier, Universitaire Ste-Justine Research Center, Montréal, QC H3T 1C5, Canada
| | - Gael Cagnone
- Departments of Pediatrics, Ophthalmology, and Pharmacology, Centre Hospitalier, Universitaire Ste-Justine Research Center, Montréal, QC H3T 1C5, Canada
| | - Yotaro Ochi
- Department of Pathology and Tumour Biology, Graduate School of Medicine, Kyoto University, Kyoto 606-8315, Japan
| | - Sergio Crespo-Garcia
- Departments of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, QC H1T 2M4, Canada
- Department of Biochemistry and Molecular Medicine, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, QC H1T 2M4, Canada
| | - Ariel M Wilson
- Departments of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, QC H1T 2M4, Canada
| | - Przemyslaw Sapieha
- Departments of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, QC H1T 2M4, Canada
- Department of Biochemistry and Molecular Medicine, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, QC H1T 2M4, Canada
| |
Collapse
|
24
|
Zhao B, Zhao Y, Sun X. Mechanism and therapeutic targets of circulating immune cells in diabetic retinopathy. Pharmacol Res 2024; 210:107505. [PMID: 39547465 DOI: 10.1016/j.phrs.2024.107505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 11/06/2024] [Accepted: 11/11/2024] [Indexed: 11/17/2024]
Abstract
Diabetic retinopathy (DR) continues to be the leading cause of preventable vision loss among working-aged adults, marked by immune dysregulation within the retinal microenvironment. Typically, the retina is considered as an immune-privileged organ, where circulating immune cells are restricted from entry under normal conditions. However, during the progression of DR, this immune privilege is compromised as circulating immune cells breach the barrier and infiltrate the retina. Increasing evidence suggests that vascular and neuronal degeneration in DR is largely driven by the infiltration of immune cells, particularly neutrophils, monocyte-derived macrophages, and lymphocytes. This review delves into the mechanisms and therapeutic targets associated with these immune cell populations in DR, offering a promising and innovative approach to managing the disease.
Collapse
Affiliation(s)
- Bowen Zhao
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yin Zhao
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Xufang Sun
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
25
|
Geraldo LH, Xu Y, Mouthon G, Furtado J, Leser FS, Blazer LL, Adams JJ, Zhang S, Zheng L, Song E, Robinson ME, Thomas JL, Sidhu SS, Eichmann A. Monoclonal antibodies that block Roundabout 1 and 2 signaling target pathological ocular neovascularization through myeloid cells. Sci Transl Med 2024; 16:eadn8388. [PMID: 39565875 PMCID: PMC11822886 DOI: 10.1126/scitranslmed.adn8388] [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: 01/02/2024] [Revised: 07/29/2024] [Accepted: 10/31/2024] [Indexed: 11/22/2024]
Abstract
Roundabout (ROBO) 1 and 2 are transmembrane receptors that bind secreted SLIT ligands through their extracellular domains (ECDs) and signal through their cytoplasmic domains to modulate the cytoskeleton and regulate cell migration, adhesion, and proliferation. SLIT-ROBO signaling regulates pathological ocular neovascularization, which is a major cause of vision loss worldwide, but pharmacological tools to prevent SLIT-ROBO signaling are lacking. Here, we developed human monoclonal antibodies (mAbs) against the ROBO1 and ROBO2 ECDs. One antibody that inhibited in vitro SLIT2 signaling through ROBO1 and ROBO2 (anti-ROBO1/2) also reduced ocular neovascularization in oxygen-induced retinopathy (OIR) and laser-induced corneal neovascularization (CNV) mouse models in vivo. Single-cell RNA sequencing of OIR retinas revealed that antibody treatment affected several cell types relevant to physiological and pathological angiogenesis, including endothelial cells, pericytes, and a heterogeneous population of myeloid cells. mAb treatment improved blood-retina barrier integrity and prevented pathological pericyte activation in OIR. SLIT-ROBO signaling inhibition prevented pathological activation of myeloid cells and increased neuroprotective myeloid populations normally seen in the developing retina. Microglia/infiltrating macrophage-specific ablation of Robo1 and Robo2 or knockout of the downstream effector phosphatidylinositol 3-kinase (Pik3cg) encoding PI3Kγ in both OIR and CNV models phenocopied anti-ROBO1/2 treatment, further demonstrating the key role of myeloid cells as drivers of ocular neovascular diseases. ROBO1/2 blocking antibodies may thus provide a promising strategy to combat inflammation in blinding eye diseases.
Collapse
Affiliation(s)
- Luiz Henrique Geraldo
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Yunling Xu
- Université de Paris, INSERM, PARCC, F-75015 Paris, France
| | - Gaspard Mouthon
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Jessica Furtado
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06510, USA
| | | | - Levi L. Blazer
- School of Pharmacy, University of Waterloo, Kitchener, Ontario N2G 1C5, Canada
| | - Jarrett J. Adams
- School of Pharmacy, University of Waterloo, Kitchener, Ontario N2G 1C5, Canada
| | - Sophia Zhang
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Lana Zheng
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Eric Song
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA
- Department of Ophthalmology and Visual Sciences, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Mark E. Robinson
- Center of Molecular and Cellular Oncology, Yale Cancer Center, Yale School of Medicine, New Haven, CT 06510, USA
| | - Jean-Leon Thomas
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA
- Paris Brain Institute, Université Pierre et Marie Curie Paris 06 UMRS1127, Sorbonne Université, F-75013 Paris, France
| | - Sachdev S. Sidhu
- School of Pharmacy, University of Waterloo, Kitchener, Ontario N2G 1C5, Canada
| | - Anne Eichmann
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06510, USA
- Université de Paris, INSERM, PARCC, F-75015 Paris, France
| |
Collapse
|
26
|
Haliyur R, Parkinson DH, Ma F, Xu J, Li Q, Huang Y, Tsoi LC, Bogle R, Liu J, Gudjonsson JE, Rao RC. Liquid Biopsy for Proliferative Diabetic Retinopathy: Single-Cell Transcriptomics of Human Vitreous Reveals Inflammatory T-Cell Signature. OPHTHALMOLOGY SCIENCE 2024; 4:100539. [PMID: 39220810 PMCID: PMC11365369 DOI: 10.1016/j.xops.2024.100539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 04/16/2024] [Accepted: 04/19/2024] [Indexed: 09/04/2024]
Abstract
Purpose Current therapies for proliferative diabetic retinopathy (PDR) do not specifically target VEGF-independent, cell-type-specific processes that lead to vision loss, such as inflammatory pathways. This study aimed to identify targetable cell types and corresponding signaling pathways by elucidating the single-cell landscape of the vitreous of patients with PDR. Design Case series. Subjects Vitreous and peripheral blood obtained from 5 adult patients (6 eyes) undergoing pars plana vitrectomy for vision-threatening PDR. Methods Single-cell RNA sequencing (scRNA-seq) was performed on vitreous cells obtained from diluted cassette washings during vitrectomy from 6 eyes and peripheral blood mononuclear cells (PBMCs, n = 5). Droplet-based scRNA-seq was performed using the Chromium 10x platform to obtain single-cell transcriptomes. Differences in tissue compartments were analyzed with gene ontology enrichment of differentially expressed genes and an unbiased ligand-receptor interaction analysis. Main Outcome Measures Single-cell transcriptomic profiles of vitreous and peripheral blood. Results Transcriptomes from 13 675 surgically harvested vitreous cells and 22 636 PBMCs were included. Clustering revealed 4 cell states consistently across all eyes with representative transcripts for T cells (CD2, CD3D, CD3E, and GZMA), B cells (CD79A, IGHM, MS4A1 (CD20), and HLA-DRA), myeloid cells (LYZ, CST3, AIF1, and IFI30), and neutrophils (BASP1, CXCR2, S100A8, and S100A9). Most vitreous cells were T cells (91.6%), unlike the peripheral blood (46.2%), whereas neutrophils in the vitreous were essentially absent. The full repertoire of adaptive T cells including CD4+, CD8+ and T regulatory cells (Treg) and innate immune system effectors (i.e., natural killer T cells) was present in the vitreous. Pathway analysis also demonstrated activation of CD4+ and CD8+ memory T cells and ligand-receptor interactions unique to the vitreous. Conclusions In the first single-cell transcriptomic characterization of human vitreous in a disease state, we show PDR vitreous is primarily composed of T cells, a critical component of adaptive immunity, with activity and proportions distinct from T cells within the peripheral blood, and neutrophils are essentially absent. These results demonstrate the feasibility of liquid vitreous biopsies via collection of otherwise discarded, diluted cassette washings during vitrectomy to gain mechanistic and therapeutic insights into human vitreoretinal disease. Financial Disclosures Proprietary or commercial disclosure may be found in the Footnotes and Disclosures at the end of this article.
Collapse
Affiliation(s)
- Rachana Haliyur
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, Michigan
| | - David H. Parkinson
- Medical Scientist Training Program, University of Michigan Medical School, University of Michigan, Ann Arbor, Michigan
| | - Feiyang Ma
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Jing Xu
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, Michigan
| | - Qiang Li
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, Michigan
| | - Yuanhao Huang
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan
| | - Lam C. Tsoi
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Rachael Bogle
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Jie Liu
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan
| | - Johann E. Gudjonsson
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Rajesh C. Rao
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, Michigan
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
- Center for RNA Biomedicine, University of Michigan, Ann Arbor, Michigan
- A. Alfred Taubman Medical Research Institute, University of Michigan, Ann Arbor, Michigan
- Section of Ophthalmology, Surgery Service, Veterans Administration Ann Arbor Healthcare System, Ann Arbor, Michigan
| |
Collapse
|
27
|
Yagi H, Boeck M, Petrishka-Lozenska M, Lundgren P, Kasai T, Cagnone G, Neilsen K, Wang C, Lee J, Tomita Y, Singh SA, Joyal JS, Aikawa M, Negishi K, Fu Z, Hellström A, Smith LEH. Timed topical dexamethasone eye drops improve mitochondrial function to prevent severe retinopathy of prematurity. Angiogenesis 2024; 27:903-917. [PMID: 39287727 PMCID: PMC11564262 DOI: 10.1007/s10456-024-09948-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 08/30/2024] [Indexed: 09/19/2024]
Abstract
Pathological neovascularization in retinopathy of prematurity (ROP) can cause visual impairment in preterm infants. Current ROP treatments which are not preventative and only address late neovascular ROP, are costly and can lead to severe complications. We showed that topical 0.1% dexamethasone eye drops administered prior to peak neovessel formation prevented neovascularization in five extremely preterm infants at high risk for ROP and suppressed neovascularization by 30% in mouse oxygen-induced retinopathy (OIR) modeling ROP. In contrast, in OIR, topical dexamethasone treatment before any neovessel formation had limited efficacy in preventing later neovascularization, while treatment after peak neovessel formation had a non-statistically significant trend to exacerbating disease. Optimally timed topical dexamethasone suppression of neovascularization in OIR was associated with increased retinal mitochondrial gene expression and decreased inflammatory marker expression, predominantly found in immune cells. Blocking mitochondrial ATP synthetase reversed the inhibitory effect of dexamethasone on neovascularization in OIR. This study provides new insights into topical steroid effects in retinal neovascularization and into mitochondrial function in phase II ROP, and suggests a simple clinical approach to prevent severe ROP.
Collapse
Affiliation(s)
- Hitomi Yagi
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, 3 Blackfan Circle, CLS 18, Boston, MA, 02115, USA
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Myriam Boeck
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, 3 Blackfan Circle, CLS 18, Boston, MA, 02115, USA
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, 79106, Germany
| | - Mariya Petrishka-Lozenska
- The Sahlgrenska Centre for Pediatric Ophthalmology Research, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, 40530, Sweden
| | - Pia Lundgren
- The Sahlgrenska Centre for Pediatric Ophthalmology Research, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, 40530, Sweden
| | - Taku Kasai
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Gael Cagnone
- CHU Sainte-Justine Research Center, Montreal, QC, CA, H3T 1C5, Canada
- Department of Pediatrics, Ophthalmology, and Pharmacology, CHU Sainte-Justine, Université de Montréal, Montreal, QC, CA, H3T 1C5, Canada
| | - Katherine Neilsen
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, 3 Blackfan Circle, CLS 18, Boston, MA, 02115, USA
| | - Chaomei Wang
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, 3 Blackfan Circle, CLS 18, Boston, MA, 02115, USA
| | - Jeff Lee
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, 3 Blackfan Circle, CLS 18, Boston, MA, 02115, USA
| | - Yohei Tomita
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, 3 Blackfan Circle, CLS 18, Boston, MA, 02115, USA
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Sasha A Singh
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Jean-Sébastien Joyal
- CHU Sainte-Justine Research Center, Montreal, QC, CA, H3T 1C5, Canada
- Department of Pediatrics, Ophthalmology, and Pharmacology, CHU Sainte-Justine, Université de Montréal, Montreal, QC, CA, H3T 1C5, Canada
- Department of Ophthalmology, Université de Montréal, Montreal, QC, CA, H3T 1J4, Canada
| | - Masanori Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Kazuno Negishi
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Zhongjie Fu
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, 3 Blackfan Circle, CLS 18, Boston, MA, 02115, USA
| | - Ann Hellström
- The Sahlgrenska Centre for Pediatric Ophthalmology Research, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, 40530, Sweden.
| | - Lois E H Smith
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, 3 Blackfan Circle, CLS 18, Boston, MA, 02115, USA.
| |
Collapse
|
28
|
Liao YL, Fang YF, Sun JX, Dou GR. Senescent endothelial cells: a potential target for diabetic retinopathy. Angiogenesis 2024; 27:663-679. [PMID: 39215875 PMCID: PMC11564237 DOI: 10.1007/s10456-024-09943-7] [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: 04/29/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Abstract
Diabetic retinopathy (DR) is a diabetic complication that results in visual impairment and relevant retinal diseases. Current therapeutic strategies on DR primarily focus on antiangiogenic therapies, which particularly target vascular endothelial growth factor and its related signaling transduction. However, these therapies still have limitations due to the intricate pathogenesis of DR. Emerging studies have shown that premature senescence of endothelial cells (ECs) in a hyperglycemic environment is involved in the disease process of DR and plays multiple roles at different stages. Moreover, these surprising discoveries have driven the development of senotherapeutics and strategies targeting senescent endothelial cells (SECs), which present challenging but promising prospects in DR treatment. In this review, we focus on the inducers and mechanisms of EC senescence in the pathogenesis of DR and summarize the current research advances in the development of senotherapeutics and strategies that target SECs for DR treatment. Herein, we highlight the role played by key factors at different stages of EC senescence, which will be critical for facilitating the development of future innovative treatment strategies that target the different stages of senescence in DR.
Collapse
Affiliation(s)
- Ying-Lu Liao
- Department of Ophthalmology, Eye Institute of Chinese PLA, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
- Department of the Cadet Team 6 of the School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Yi-Fan Fang
- Department of Ophthalmology, Eye Institute of Chinese PLA, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Jia-Xing Sun
- Department of Ophthalmology, Eye Institute of Chinese PLA, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Guo-Rui Dou
- Department of Ophthalmology, Eye Institute of Chinese PLA, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
29
|
Sharma S, Cheema M, Reeson PL, Narayana K, Boghozian R, Cota AP, Brosschot TP, FitzPatrick RD, Körbelin J, Reynolds LA, Brown CE. A pathogenic role for IL-10 signalling in capillary stalling and cognitive impairment in type 1 diabetes. Nat Metab 2024; 6:2082-2099. [PMID: 39496927 PMCID: PMC11599051 DOI: 10.1038/s42255-024-01159-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 10/04/2024] [Indexed: 11/06/2024]
Abstract
Vascular pathology is associated with cognitive impairment in diseases such as type 1 diabetes; however, how capillary flow is affected and the underlying mechanisms remain elusive. Here we show that capillaries in the diabetic mouse brain in both sexes are prone to stalling, with blocks consisting primarily of erythrocytes in branches off ascending venules. Screening for circulating inflammatory cytokines revealed persistently high levels of interleukin-10 (IL-10) in diabetic mice. Contrary to expectation, stimulating IL-10 signalling increased capillary obstruction, whereas inhibiting IL-10 receptors with neutralizing antibodies or endothelial specific knockdown in diabetic mice reversed these impairments. Chronic treatment of diabetic mice with IL-10 receptor neutralizing antibodies improved cerebral blood flow, increased capillary flux and diameter, downregulated haemostasis and cell adhesion-related gene expression, and reversed cognitive deficits. These data suggest that IL-10 signalling has an unexpected pathogenic role in cerebral microcirculatory defects and cognitive impairment associated with type 1 diabetes.
Collapse
Affiliation(s)
- Sorabh Sharma
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Manjinder Cheema
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Patrick L Reeson
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Kamal Narayana
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Roobina Boghozian
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Ana Paula Cota
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Tara P Brosschot
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada
| | - Rachael D FitzPatrick
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada
| | - Jakob Körbelin
- Department of Oncology, Hematology and Bone Marrow Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lisa A Reynolds
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada
| | - Craig E Brown
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada.
- Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
30
|
Lu C, Mao X, Yuan S. Decoding physiological and pathological roles of innate immune cells in eye diseases: the perspectives from single-cell RNA sequencing. Front Immunol 2024; 15:1490719. [PMID: 39544948 PMCID: PMC11560449 DOI: 10.3389/fimmu.2024.1490719] [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: 09/03/2024] [Accepted: 10/09/2024] [Indexed: 11/17/2024] Open
Abstract
Single-cell RNA sequencing (scRNA-seq) has facilitated a deeper comprehension of the molecular mechanisms behind eye diseases and has prompted the selection of precise therapeutic targets by examining the cellular and molecular intricacies at the single-cell level. This review delineates the pivotal role of scRNA-seq in elucidating the functions of innate immune cells within the context of ocular pathologies. Recent advancements in scRNA-seq have revealed that innate immune cells, both from the periphery and resident in the retina, are actively engaged in various stages of multiple eye diseases. Notably, resident microglia and infiltrating neutrophils exhibit swift responses during the initial phase of injury, while peripheral monocyte-derived macrophages exhibit transcriptomic profiles akin to those of activated microglia, suggesting their potential for long-term residence within the retina. The scRNA-seq analyses have underscored the cellular heterogeneity and gene expression alterations within innate immune cells, which, while sharing commonalities, exhibit disease-specific variations. These insights have not only broadened our understanding of the cellular and molecular mechanisms in eye diseases but also paved the way for the identification of candidate targets for targeted therapeutic interventions. The application of scRNA-seq technology has heralded a new era in the study of ocular pathologies, enabling a more detailed appreciation of the roles that innate immune cells play across a spectrum of eye diseases.
Collapse
Affiliation(s)
- Chen Lu
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiying Mao
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Songtao Yuan
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
31
|
Chen L, Zhang H, Zhang Y, Li X, Wang M, Shen Y, Cao Y, Xu Y, Yao J. Ganglion cell-derived LysoPS induces retinal neovascularisation by activating the microglial GPR34-PI3K-AKT-NINJ1 axis. J Neuroinflammation 2024; 21:278. [PMID: 39468551 PMCID: PMC11520652 DOI: 10.1186/s12974-024-03265-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 10/18/2024] [Indexed: 10/30/2024] Open
Abstract
Retinal neovascularisation is a major cause of blindness in patients with proliferative diabetic retinopathy (PDR). It is mediated by the complex interaction between dysfunctional ganglion cells, microglia, and vascular endothelial cells. Notably, retinal microglia, the intrinsic immune cells of the retina, play a crucial role in the pathogenesis of retinopathy. In this study, we found that lysophosphatidylserines (LysoPS) released from injured ganglion cells induced microglial extracellular trap formation and retinal neovascularisation. Mechanistically, LysoPS activated the GPR34-PI3K-AKT-NINJ1 signalling axis by interacting with the GPR34 receptor on the microglia. This activation upregulated the expression of inflammatory cytokines, such as IL-6, IL-8, VEGFA, and FGF2, and facilitated retinal vascular endothelial cell angiogenesis. As a result, inhibition of the GPR34-PI3K-AKT-NINJ1 axis significantly decreased microglial extracellular trap formation and neovascularisation by suppressing LysoPS-induced microglial inflammatory responses, both in vitro and in vivo. This study reveals the crucial role of apoptotic ganglion cells in activating microglial inflammation in PDR, thereby enhancing our understanding of the pathogenesis of retinal neovascularisation.
Collapse
Affiliation(s)
- Lushu Chen
- The Affiliated Eye Hospital, Nanjing Medical University, 138 Hanzhong Road, Nanjing, 210029, P. R. China
- The Fourth School of Clinical Medicine, Nanjing Medical University, 140 Hanzhong Road, Nanjing, 210029, P. R. China
| | - HuiYing Zhang
- The Affiliated Eye Hospital, Nanjing Medical University, 138 Hanzhong Road, Nanjing, 210029, P. R. China
- The Fourth School of Clinical Medicine, Nanjing Medical University, 140 Hanzhong Road, Nanjing, 210029, P. R. China
| | - Ying Zhang
- The Affiliated Eye Hospital, Nanjing Medical University, 138 Hanzhong Road, Nanjing, 210029, P. R. China
- The Fourth School of Clinical Medicine, Nanjing Medical University, 140 Hanzhong Road, Nanjing, 210029, P. R. China
| | - Xiumiao Li
- The Affiliated Eye Hospital, Nanjing Medical University, 138 Hanzhong Road, Nanjing, 210029, P. R. China
| | - MeiHuan Wang
- The Affiliated Eye Hospital, Nanjing Medical University, 138 Hanzhong Road, Nanjing, 210029, P. R. China
| | - Yaming Shen
- The Affiliated Eye Hospital, Nanjing Medical University, 138 Hanzhong Road, Nanjing, 210029, P. R. China
| | - Yuan Cao
- The Fourth School of Clinical Medicine, Nanjing Medical University, 140 Hanzhong Road, Nanjing, 210029, P. R. China
| | - Yong Xu
- The Affiliated Eye Hospital, Nanjing Medical University, 138 Hanzhong Road, Nanjing, 210029, P. R. China.
- The Fourth School of Clinical Medicine, Nanjing Medical University, 140 Hanzhong Road, Nanjing, 210029, P. R. China.
| | - Jin Yao
- The Affiliated Eye Hospital, Nanjing Medical University, 138 Hanzhong Road, Nanjing, 210029, P. R. China.
- The Fourth School of Clinical Medicine, Nanjing Medical University, 140 Hanzhong Road, Nanjing, 210029, P. R. China.
| |
Collapse
|
32
|
Liu J, Guo B, Liu Q, Zhu G, Wang Y, Wang N, Yang Y, Fu S. Cellular Senescence: A Bridge Between Diabetes and Microangiopathy. Biomolecules 2024; 14:1361. [PMID: 39595537 PMCID: PMC11591988 DOI: 10.3390/biom14111361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/21/2024] [Accepted: 10/24/2024] [Indexed: 11/28/2024] Open
Abstract
Cellular senescence is a state of permanent cell cycle arrest and plays an important role in many vascular lesions. This study found that the cells of diabetic patients have more characteristics of senescence, which may cause microvascular complications. Cell senescence, as one of the common fates of cells, links microangiopathy and diabetes. Cell senescence in a high-glucose environment can partially elucidate the mechanism of diabetic microangiopathy, and various types of cellular senescence induced by it can promote the progression of diabetic microangiopathy. Still, the molecular mechanism of microangiopathy-related cellular senescence has not yet been clearly studied. Building on recent research evidence, we herein summarize the fundamental mechanisms underlying the development of cellular senescence in various microangiopathies associated with diabetes. We gradually explain how cellular senescence serves as a key driver of diabetic microangiopathy. At the same time, the treatment of basic senescence mechanisms such as cellular senescence may have a great impact on the pathogenesis of the disease, may be more effective in preventing the development of diabetic microangiopathy, and may provide new ideas for the clinical treatment and prognosis of diabetic microangiopathy.
Collapse
Affiliation(s)
- Jiahui Liu
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China; (J.L.); (B.G.); (Q.L.); (G.Z.); (Y.W.); (N.W.); (Y.Y.)
| | - Buyu Guo
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China; (J.L.); (B.G.); (Q.L.); (G.Z.); (Y.W.); (N.W.); (Y.Y.)
| | - Qianqian Liu
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China; (J.L.); (B.G.); (Q.L.); (G.Z.); (Y.W.); (N.W.); (Y.Y.)
| | - Guomao Zhu
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China; (J.L.); (B.G.); (Q.L.); (G.Z.); (Y.W.); (N.W.); (Y.Y.)
| | - Yaqi Wang
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China; (J.L.); (B.G.); (Q.L.); (G.Z.); (Y.W.); (N.W.); (Y.Y.)
| | - Na Wang
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China; (J.L.); (B.G.); (Q.L.); (G.Z.); (Y.W.); (N.W.); (Y.Y.)
| | - Yichen Yang
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China; (J.L.); (B.G.); (Q.L.); (G.Z.); (Y.W.); (N.W.); (Y.Y.)
| | - Songbo Fu
- Department of Endocrinology, The First Hospital of Lanzhou University, Lanzhou 730000, China
- Gansu Province Clinical Research Center for Endocrine Disease, Lanzhou 730000, China
| |
Collapse
|
33
|
Hao L, Wang S, Zhang L, Huang J, Zhang Y, Qin X. Transcriptome sequencing and Mendelian randomization analysis identified biomarkers related to neutrophil extracellular traps in diabetic retinopathy. Front Immunol 2024; 15:1408974. [PMID: 39483475 PMCID: PMC11524841 DOI: 10.3389/fimmu.2024.1408974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 09/27/2024] [Indexed: 11/03/2024] Open
Abstract
Summary In the development of diabetic retinopathy (DR), neutrophil infiltration hastens the adhesion between neutrophils and endothelial cells, leading to inflammation. Meanwhile, neutrophil extracellular traps (NETs) produced by neutrophils could clear aging blood vessels, setting the stage for retinal vascular regeneration. To explore the mechanism of NETs-related genes in DR, the transcriptome of NETs from normal and DR individuals were analyzed with gene sequencing and mendelian randomization (MR) analysis. Five NETs-related genes were identified as key genes. Among these genes, CLIC3, GBP2, and P2RY12 were found to be risk factors for Proliferative DR(PDR), whereas HOXA1 and PSAP were protective factors. Further verification by qRT-PCR recognized GBP2, P2RY12 and PSAP as NETs-associated biomarkers in PDR. Purpose To investigate neutrophil extracellular traps (NETs) related genes as biomarkers in the progression of diabetic retinopathy (DR). Methods We collected whole blood samples from 10 individuals with DR and 10 normal controls (NCs) for transcriptome sequencing. Following quality control and preprocessing of the sequencing data, differential expression analysis was conducted to identify differentially expressed genes (DEGs) between the DR and NC groups. Candidate genes were then selected by intersecting these DEGs with key module genes identified through weighted gene co-expression network analysis. These candidate genes were subjected to mendelian randomization (MR) analysis, then least absolute shrinkage and selection operator analysis to pinpoint key genes. The diagnostic utility of these key genes was evaluated using receiver operating characteristic curve analysis, and their expression levels were examined. Additional analysis, including nomogram construction, gene set enrichment analysis, drug prediction and molecular docking, were performed to investigate the functions and molecular mechanisms of the key genes. Finally, the expression of key genes was verified by qRT-PCR and biomarkers were identified. Results Intersection of 1,004 DEGs with 1,038 key module genes yielded 291 candidate genes. Five key genes were identified: HOXA1, GBP2, P2RY12, CLIC3 and PSAP. Among them, CLIC3, GBP2, and P2RY12 were identified as risk factors for DR, while HOXA1 and PSAP were protective. These key genes demonstrated strong diagnostic performance for DR. With the exception of P2RY12, all other key genes exhibited down-regulation in the DR group. Furthermore, the nomogram incorporating multiple key genes demonstrated superior predictive capacity for DR compared to a single key genes. The identified key genes are involved in oxidative phosphorylation and ribosome functions. Drug predictions targeting P2RY12 suggested prasugrel, ticagrelor, and ticlopidine as potential options owing to their high binding affinity with this key genes. The qRT-PCR results revealed that the results of GBP2, PSAP and P2RY12 exhibited consistent expression patterns with the dataset. Conclusion This study identified GBP2, P2RY12 and PSAP as NETs-associated biomarkers in the development of PDR, offering new insights for clinical diagnosis and potential treatment strategies for DR.
Collapse
Affiliation(s)
- Linlin Hao
- Department of Ophthalmology, the Second Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Songhong Wang
- Department of Ophthalmology, the Second Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lian Zhang
- Department of Ophthalmology, the Second Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, China
- Affiliated Eye Hospital of Shandong University of Traditional Chinese Medicine, Shandong University, Jinan, China
| | - Jie Huang
- Affiliated Eye Hospital of Shandong University of Traditional Chinese Medicine, Shandong University, Jinan, China
| | - Yue Zhang
- Department of Operating Room, the Second Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xuejiao Qin
- Department of Ophthalmology, the Second Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
34
|
Chen S, Liu Y, Zhang Y, Guo X, Bai T, He K, Zhu Y, Lei Y, Du M, Wang X, Liu Q, Yan H. Bruton's tyrosine kinase inhibition suppresses pathological retinal angiogenesis. Br J Pharmacol 2024. [PMID: 39374939 DOI: 10.1111/bph.17344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 06/25/2024] [Accepted: 08/22/2024] [Indexed: 10/09/2024] Open
Abstract
BACKGROUND AND PURPOSE Pathological retinal angiogenesis is a typical manifestation of vision-threatening ocular diseases. Many patients exhibit poor response or resistance to anti-vascular endothelial growth factor (VEGF) agents. Bruton's tyrosine kinase (BTK) controls the proliferation and function of immune cells. Therefore, we examined the anti-inflammatory and anti-angiogenic effects of BTK inhibition on retinal angiogenesis. EXPERIMENTAL APPROACH Retinal neovascularisation and vascular leakage in oxygen-induced retinopathy in C57/BL6J mice were assessed by whole-mount retinal immunofluorescence. PLX5622 was used to deplete microglia and Rag1-knockout mice were used to test the contribution of lymphocytes to the effects of BTK inhibition. The cytokines, activation markers, inflammatory and immune-regulatory activities of retinal microglia/macrophages were detected using qRT-PCR and immunofluorescence. NLRP3 was detected by western blotting, and the effects of BTK inhibition on the co-culture of microglia and human retinal microvascular endothelial cells (HRMECs) were examined. KEY RESULTS BTK inhibition suppressed pathological angiogenesis and vascular leakage, and significantly reduced retinal inflammation, which involved microglia/macrophages but not lymphocytes. BTK inhibition increased anti-inflammatory factors and reduced pro-inflammatory cytokines that resulted from NLRP3 inflammasome activation. BTK inhibition suppressed the inflammatory activity of microglia/macrophages, and acted synergistically with anti-VEGF without retinal toxicity. Moreover, the supernatant of microglia incubated with BTK-inhibitor reduced the proliferation, tube formation and sprouting of HRMECs. CONCLUSION AND IMPLICATIONS BTK inhibition suppressed retinal neovascularisation and vascular leakage by modulating the inflammatory activity of microglia and macrophages. Our study suggests BTK inhibition as a novel and promising approach for alleviating pathological retinal angiogenesis.
Collapse
Affiliation(s)
- Siyue Chen
- Department of Ophthalmology, Tianjin Medical University General Hospital, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Key Laboratory of Ocular Trauma, Tianjin Institute of Eye Health and Eye Diseases, China-UK "Belt and Road" Ophthalmology Joint Laboratory, Tianjin, China
| | - Yuming Liu
- Department of Ophthalmology, Tianjin Medical University General Hospital, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Key Laboratory of Ocular Trauma, Tianjin Institute of Eye Health and Eye Diseases, China-UK "Belt and Road" Ophthalmology Joint Laboratory, Tianjin, China
| | - Yutian Zhang
- Department of Ophthalmology, Tianjin Medical University General Hospital, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Key Laboratory of Ocular Trauma, Tianjin Institute of Eye Health and Eye Diseases, China-UK "Belt and Road" Ophthalmology Joint Laboratory, Tianjin, China
| | - Xu Guo
- Department of Ophthalmology, Tianjin Medical University General Hospital, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Key Laboratory of Ocular Trauma, Tianjin Institute of Eye Health and Eye Diseases, China-UK "Belt and Road" Ophthalmology Joint Laboratory, Tianjin, China
| | - Tinghui Bai
- Department of Ophthalmology, Tianjin Medical University General Hospital, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Key Laboratory of Ocular Trauma, Tianjin Institute of Eye Health and Eye Diseases, China-UK "Belt and Road" Ophthalmology Joint Laboratory, Tianjin, China
| | - Kai He
- Department of Ophthalmology, Tianjin Medical University General Hospital, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Key Laboratory of Ocular Trauma, Tianjin Institute of Eye Health and Eye Diseases, China-UK "Belt and Road" Ophthalmology Joint Laboratory, Tianjin, China
| | - Yanfang Zhu
- Department of Ophthalmology, Tianjin Medical University General Hospital, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Key Laboratory of Ocular Trauma, Tianjin Institute of Eye Health and Eye Diseases, China-UK "Belt and Road" Ophthalmology Joint Laboratory, Tianjin, China
| | - Yi Lei
- Department of Ophthalmology, Tianjin Medical University General Hospital, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Key Laboratory of Ocular Trauma, Tianjin Institute of Eye Health and Eye Diseases, China-UK "Belt and Road" Ophthalmology Joint Laboratory, Tianjin, China
| | - Mei Du
- Department of Ophthalmology, Tianjin Medical University General Hospital, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Key Laboratory of Ocular Trauma, Tianjin Institute of Eye Health and Eye Diseases, China-UK "Belt and Road" Ophthalmology Joint Laboratory, Tianjin, China
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, the Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Xiaohong Wang
- Department of Ophthalmology, Tianjin Medical University General Hospital, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Key Laboratory of Ocular Trauma, Tianjin Institute of Eye Health and Eye Diseases, China-UK "Belt and Road" Ophthalmology Joint Laboratory, Tianjin, China
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, the Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Qiang Liu
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Hua Yan
- Department of Ophthalmology, Tianjin Medical University General Hospital, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Key Laboratory of Ocular Trauma, Tianjin Institute of Eye Health and Eye Diseases, China-UK "Belt and Road" Ophthalmology Joint Laboratory, Tianjin, China
| |
Collapse
|
35
|
Gao L, Peng L, Tang H, Wang C, Wang Q, Luo Y, Chen W, Xia Y. Screening and identification of differential-expressed RNAs in thrombin-induced in vitro model of intracerebral hemorrhage. Mol Cell Biochem 2024; 479:2755-2767. [PMID: 37943469 DOI: 10.1007/s11010-023-04879-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 10/07/2023] [Indexed: 11/10/2023]
Abstract
Survival of olfactory mucosal mesenchymal stem cells (OM-MSCs) remains the low level in the cerebral microenvironment during intracerebral hemorrhage (ICH). This article aims to reveal the differential expression profile of ICH-stimulated OM-MSCs based on whole transcriptome sequence analysis. OM-MSCs were isolated from 6-week C57BL/6 mice. Morphology and surface markers of OM-MSCs were investigated by light microscope and flow cytometry, respectively. OM-MSCs were incubated with 20 U/mL thrombin for 24 h to mimic ICH-induced injury in vitro. Total RNA was extracted for whole transcriptome sequencing and qPCR. OM-MSCs were characterized by negative for CD45 and CD34, and positive for CD44, CD90 and CD29. Thrombin led to decrease in cell viability and increase in senescence and apoptosis in OM-MSCs. In total, 736 lncRNAs (upregulated: 393; downregulated: 343), 21 miRNAs (upregulated: 7; downregulated: 14) and 807 mRNAs (upregulated: 422; downregulated: 385) were identified. GO and KEGG pathways were enriched in protein heterodimerization activity, trans-synaptic signaling, membrane pathway, alcohol metabolic process, organic hydroxy compound biosynthesis process, secondary alcohol metabolic process, alcoholism, neutrophil extracellular trap formation, systemic lupus erythematosus, metabolic process, steroid biosynthesis and drug metabolism-cytochrome P450. 200 lncRNA-miRNA-mRNA were predicted in thrombin-induced OM-MSCs. Based on qPCR, we validated COMMD1B, MOAP1, lncRNA CAPN15, lncRNA ALDH1L2, miR-3473b and miR-1964-3p were upregulated in thrombin-stimulated OM-MSCs, and GM20431, lncRNA GAPDH and miR-122b-3p were downregulated. Our findings provide novel understanding for thrombin-induced injury in OM-MSCs. Differently-expressed RNAs can be the targets of improving therapeutic application of OM-MSCs.
Collapse
Affiliation(s)
- Ling Gao
- Department of Neurosurgery, Affiliated Haikou Hospital, Xiangya School of Medicine, Central South University, 43 Renmin Avenue, Haikou, 570208, Hainan, China
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA, 92354, USA
| | - Li Peng
- Department of Ophthalmology, Affiliated Haikou Hospital, Xiangya School of Medicine, Central South University, 43 Renmin Avenue, Haikou, 570208, Hainan, China
| | - Hong Tang
- Department of Neurosurgery, Affiliated Haikou Hospital, Xiangya School of Medicine, Central South University, 43 Renmin Avenue, Haikou, 570208, Hainan, China
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA, 92354, USA
| | - Chuang Wang
- Department of Neurosurgery, Affiliated Haikou Hospital, Xiangya School of Medicine, Central South University, 43 Renmin Avenue, Haikou, 570208, Hainan, China
| | - Qingsong Wang
- Department of Neurosurgery, Affiliated Haikou Hospital, Xiangya School of Medicine, Central South University, 43 Renmin Avenue, Haikou, 570208, Hainan, China
| | - Yujie Luo
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA, 92354, USA
| | - Weiming Chen
- Department of Neurosurgery, Affiliated Haikou Hospital, Xiangya School of Medicine, Central South University, 43 Renmin Avenue, Haikou, 570208, Hainan, China.
| | - Ying Xia
- Department of Neurosurgery, Affiliated Haikou Hospital, Xiangya School of Medicine, Central South University, 43 Renmin Avenue, Haikou, 570208, Hainan, China.
| |
Collapse
|
36
|
Li Y, Liu W, Wang Y, Liu T, Feng Y. Nanotechnology-Mediated Immunomodulation Strategy for Inflammation Resolution. Adv Healthc Mater 2024; 13:e2401384. [PMID: 39039994 DOI: 10.1002/adhm.202401384] [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: 04/16/2024] [Revised: 07/02/2024] [Indexed: 07/24/2024]
Abstract
Inflammation serves as a common characteristic across a wide range of diseases and plays a vital role in maintaining homeostasis. Inflammation can lead to tissue damage and the onset of inflammatory diseases. Although significant progress is made in anti-inflammation in recent years, the current clinical approaches mainly rely on the systemic administration of corticosteroids and antibiotics, which only provide short-term relief. Recently, immunomodulatory approaches have emerged as promising strategies for facilitating the resolution of inflammation. Especially, the advanced nanosystems with unique biocompatibility and multifunctionality have provided an ideal platform for immunomodulation. In this review, the pathophysiology of inflammation and current therapeutic strategies are summarized. It is mainly focused on the nanomedicines that modulate the inflammatory signaling pathways, inflammatory cells, oxidative stress, and inflammation targeting. Finally, the challenges and opportunities of nanomaterials in addressing inflammation are also discussed. The nanotechnology-mediated immunomodulation will open a new treatment strategy for inflammation therapy.
Collapse
Affiliation(s)
- Ying Li
- School of Chemical Engineering and Technology, Tianjin University, Yaguan Road 135, Tianjin, 300350, P. R. China
| | - Wen Liu
- School of Chemical Engineering and Technology, Tianjin University, Yaguan Road 135, Tianjin, 300350, P. R. China
| | - Yuanchao Wang
- School of Chemical Engineering and Technology, Tianjin University, Yaguan Road 135, Tianjin, 300350, P. R. China
| | - Taotao Liu
- Department of Gastroenterology and Hepatology, Characteristic Medical Center of the Chinese People's Armed Police Force, Tianjin Key Laboratory of Hepatopancreatic Fibrosis and Molecular Diagnosis & Treatment, Tianjin, 300162, China
| | - Yakai Feng
- School of Chemical Engineering and Technology, Tianjin University, Yaguan Road 135, Tianjin, 300350, P. R. China
- Frontiers Science Center for Synthetic Biology, Tianjin University, Weijin Road 92, Tianjin, 300072, P. R. China
- Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Weijin Road 92, Tianjin, 300072, P. R. China
| |
Collapse
|
37
|
Wang H, Kim SJ, Lei Y, Wang S, Wang H, Huang H, Zhang H, Tsung A. Neutrophil extracellular traps in homeostasis and disease. Signal Transduct Target Ther 2024; 9:235. [PMID: 39300084 PMCID: PMC11415080 DOI: 10.1038/s41392-024-01933-x] [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: 02/24/2024] [Revised: 06/25/2024] [Accepted: 07/16/2024] [Indexed: 09/22/2024] Open
Abstract
Neutrophil extracellular traps (NETs), crucial in immune defense mechanisms, are renowned for their propensity to expel decondensed chromatin embedded with inflammatory proteins. Our comprehension of NETs in pathogen clearance, immune regulation and disease pathogenesis, has grown significantly in recent years. NETs are not only pivotal in the context of infections but also exhibit significant involvement in sterile inflammation. Evidence suggests that excessive accumulation of NETs can result in vessel occlusion, tissue damage, and prolonged inflammatory responses, thereby contributing to the progression and exacerbation of various pathological states. Nevertheless, NETs exhibit dual functionalities in certain pathological contexts. While NETs may act as autoantigens, aggregated NET complexes can function as inflammatory mediators by degrading proinflammatory cytokines and chemokines. The delineation of molecules and signaling pathways governing NET formation aids in refining our appreciation of NETs' role in immune homeostasis, inflammation, autoimmune diseases, metabolic dysregulation, and cancer. In this comprehensive review, we delve into the multifaceted roles of NETs in both homeostasis and disease, whilst discussing their potential as therapeutic targets. Our aim is to enhance the understanding of the intricate functions of NETs across the spectrum from physiology to pathology.
Collapse
Affiliation(s)
- Han Wang
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Susan J Kim
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Yu Lei
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shuhui Wang
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hui Wang
- Department of Medical Genetics, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hai Huang
- Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Hongji Zhang
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, USA.
| | - Allan Tsung
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
38
|
Jia J, Wang Y, Li M, Wang F, Peng Y, Hu J, Li Z, Bian Z, Yang S. Neutrophils in the premetastatic niche: key functions and therapeutic directions. Mol Cancer 2024; 23:200. [PMID: 39277750 PMCID: PMC11401288 DOI: 10.1186/s12943-024-02107-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 09/02/2024] [Indexed: 09/17/2024] Open
Abstract
Metastasis has been one of the primary reasons for the high mortality rates associated with tumours in recent years, rendering the treatment of current malignancies challenging and representing a significant cause of recurrence in patients who have undergone surgical tumour resection. Halting tumour metastasis has become an essential goal for achieving favourable prognoses following cancer treatment. In recent years, increasing clarity in understanding the mechanisms underlying metastasis has been achieved. The concept of premetastatic niches has gained widespread acceptance, which posits that tumour cells establish a unique microenvironment at distant sites prior to their migration, facilitating their settlement and growth at those locations. Neutrophils serve as crucial constituents of the premetastatic niche, actively shaping its microenvironmental characteristics, which include immunosuppression, inflammation, angiogenesis and extracellular matrix remodelling. These characteristics are intimately associated with the successful engraftment and subsequent progression of tumour cells. As our understanding of the role and significance of neutrophils in the premetastatic niche deepens, leveraging the presence of neutrophils within the premetastatic niche has gradually attracted the interest of researchers as a potential therapeutic target. The focal point of this review revolves around elucidating the involvement of neutrophils in the formation and shaping of the premetastatic niche (PMN), alongside the introduction of emerging therapeutic approaches aimed at impeding cancer metastasis.
Collapse
Affiliation(s)
- Jiachi Jia
- Zhengzhou University, Zhengzhou, 450000, China
| | - Yuhang Wang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
| | - Mengjia Li
- Department of Haematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
| | - Fuqi Wang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
| | - Yingnan Peng
- Department of Haematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
| | - Junhong Hu
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
| | - Zhen Li
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China.
| | - Zhilei Bian
- Department of Haematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China.
| | - Shuaixi Yang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China.
| |
Collapse
|
39
|
Zhang M, Li S, Ying J, Qu Y. Neutrophils: a key component in ECMO-related acute organ injury. Front Immunol 2024; 15:1432018. [PMID: 39346902 PMCID: PMC11427252 DOI: 10.3389/fimmu.2024.1432018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 08/27/2024] [Indexed: 10/01/2024] Open
Abstract
Extracorporeal membrane oxygenation (ECMO), as an extracorporeal life support technique, can save the lives of reversible critically ill patients when conventional treatments fail. However, ECMO-related acute organ injury is a common complication that increases the risk of death in critically ill patients, including acute kidney injury, acute brain injury, acute lung injury, and so on. In ECMO supported patients, an increasing number of studies have shown that activation of the inflammatory response plays an important role in the development of acute organ injury. Cross-cascade activation of the complement system, the contact system, and the coagulation system, as well as the mechanical forces of the circuitry are very important pathophysiological mechanisms, likely leading to neutrophil activation and the production of neutrophil extracellular traps (NETs). NETs may have the potential to cause organ damage, generating interest in their study as potential therapeutic targets for ECMO-related acute organ injury. Therefore, this article comprehensively summarized the mechanism of neutrophils activation and NETs formation following ECMO treatment and their actions on acute organ injury.
Collapse
Affiliation(s)
- Mingfu Zhang
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Chronobiology (National Health Commission), West China Second University Hospital, Sichuan University, Chengdu, China
| | - Shiping Li
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Chronobiology (National Health Commission), West China Second University Hospital, Sichuan University, Chengdu, China
| | - Junjie Ying
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Chronobiology (National Health Commission), West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yi Qu
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Chronobiology (National Health Commission), West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
40
|
Wu X, Zhong L, Yu J, Wang N, Bu S, Wang H, Zhang J, Luo X, Liu Y, Nie C. MDSCs promote pathological angiogenesis in ocular neovascular disease. Biomed Pharmacother 2024; 178:117222. [PMID: 39088968 DOI: 10.1016/j.biopha.2024.117222] [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: 04/28/2024] [Revised: 07/25/2024] [Accepted: 07/26/2024] [Indexed: 08/03/2024] Open
Abstract
BACKGROUND Ocular neovascular diseases, which contribute significantly to vision loss, lack effective preventive treatments. Recent studies have highlighted the significant involvement of immune cells in neovascular retinopathy. Myeloid-derived suppressor cells (MDSCs) promote the development of neovascularization, but it is unknown whether they participate in pathological neovascularization and whether they are expected to be a therapeutic target. METHOD We investigated the role of MDSCs in promoting pathological angiogenesis using an oxygen-induced retinopathy (OIR) model, employing flow cytometry, immunofluorescence, and smart-seq analysis. Then, we evaluated the proportion of MDSCs in patient blood samples using flow cytometry. Additionally, we assessed the effect of MDSC depletion using an anti-Gr-1 monoclonal antibody on retinal vasculopathy and alterations in retinal microglia. RESULTS In the OIR model, an elevated ratio of MDSCs was observed in both blood and retinal tissue during phase II (Neovascularization). The depletion of MDSCs resulted in reduced retinal neovascularization and vaso-obliteration, along with a decrease in microglia within the neovascularization area. Furthermore, analysis of gene transcripts associated with MDSCs indicated activation of vascular endothelial growth factor (VEGF) regulation and inflammation. Importantly, infants with ROP exhibited a higher proportion of MDSCs in their blood samples. CONCLUSION Our results suggested that excessive MDSCs represent an unrecognized feature of ocular neovascular diseases and be responsible for the retinal vascular inflammation and angiogenesis, providing opportunities for new therapeutic approaches to ocular neovascular disease.
Collapse
Affiliation(s)
- Xiaojun Wu
- Neonatology Department, Guangdong Women and Children Hospital, Guangzhou, Guangdong 510000, China; National Key Clinical Specialty Construction Project/Guangdong Neonatal ICU Medical Quality Control Center, Guangzhou, Guangdong 510000, China; Center for Medical Research on Innovation and Translation, Guangzhou First People's Hospital, the Second Affiliated Hospital of South China University of Technology, Guangzhou, Guangdong 510005, China
| | - Limei Zhong
- Department of Laboratory Medicine, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong 510317, China
| | - Jun Yu
- Center for Medical Research on Innovation and Translation, Guangzhou First People's Hospital, the Second Affiliated Hospital of South China University of Technology, Guangzhou, Guangdong 510005, China
| | - Ning Wang
- Neonatology Department, Guangdong Women and Children Hospital, Guangzhou, Guangdong 510000, China; National Key Clinical Specialty Construction Project/Guangdong Neonatal ICU Medical Quality Control Center, Guangzhou, Guangdong 510000, China
| | - Shimiao Bu
- Center for Medical Research on Innovation and Translation, Guangzhou First People's Hospital, the Second Affiliated Hospital of South China University of Technology, Guangzhou, Guangdong 510005, China
| | - Huijuan Wang
- Center for Medical Research on Innovation and Translation, Guangzhou First People's Hospital, the Second Affiliated Hospital of South China University of Technology, Guangzhou, Guangdong 510005, China
| | - Jie Zhang
- Department of Rehabilitation, Guangdong Women and Children Hospital, Guangzhou, Guangdong 510000, China
| | - Xianqiong Luo
- Neonatology Department, Guangdong Women and Children Hospital, Guangzhou, Guangdong 510000, China; National Key Clinical Specialty Construction Project/Guangdong Neonatal ICU Medical Quality Control Center, Guangzhou, Guangdong 510000, China
| | - Yufeng Liu
- Center for Medical Research on Innovation and Translation, Guangzhou First People's Hospital, the Second Affiliated Hospital of South China University of Technology, Guangzhou, Guangdong 510005, China.
| | - Chuan Nie
- Neonatology Department, Guangdong Women and Children Hospital, Guangzhou, Guangdong 510000, China; National Key Clinical Specialty Construction Project/Guangdong Neonatal ICU Medical Quality Control Center, Guangzhou, Guangdong 510000, China.
| |
Collapse
|
41
|
Rolas L, Stein M, Barkaway A, Reglero-Real N, Sciacca E, Yaseen M, Wang H, Vazquez-Martinez L, Golding M, Blacksell IA, Giblin MJ, Jaworska E, Bishop CL, Voisin MB, Gaston-Massuet C, Fossati-Jimack L, Pitzalis C, Cooper D, Nightingale TD, Lopez-Otin C, Lewis MJ, Nourshargh S. Senescent endothelial cells promote pathogenic neutrophil trafficking in inflamed tissues. EMBO Rep 2024; 25:3842-3869. [PMID: 38918502 PMCID: PMC11387759 DOI: 10.1038/s44319-024-00182-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 05/28/2024] [Accepted: 06/07/2024] [Indexed: 06/27/2024] Open
Abstract
Cellular senescence is a hallmark of advanced age and a major instigator of numerous inflammatory pathologies. While endothelial cell (EC) senescence is aligned with defective vascular functionality, its impact on fundamental inflammatory responses in vivo at single-cell level remain unclear. To directly investigate the role of EC senescence on dynamics of neutrophil-venular wall interactions, we applied high resolution confocal intravital microscopy to inflamed tissues of an EC-specific progeroid mouse model, characterized by profound indicators of EC senescence. Progerin-expressing ECs supported prolonged neutrophil adhesion and crawling in a cell autonomous manner that additionally mediated neutrophil-dependent microvascular leakage. Transcriptomic and immunofluorescence analysis of inflamed tissues identified elevated levels of EC CXCL1 on progerin-expressing ECs and functional blockade of CXCL1 suppressed the dysregulated neutrophil responses elicited by senescent ECs. Similarly, cultured progerin-expressing human ECs exhibited a senescent phenotype, were pro-inflammatory and prompted increased neutrophil attachment and activation. Collectively, our findings support the concept that senescent ECs drive excessive inflammation and provide new insights into the mode, dynamics, and mechanisms of this response at single-cell level.
Collapse
Affiliation(s)
- Loïc Rolas
- Centre for Microvascular Research, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Monja Stein
- Centre for Microvascular Research, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Anna Barkaway
- Centre for Microvascular Research, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Natalia Reglero-Real
- Centre for Microvascular Research, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Elisabetta Sciacca
- Centre for Translational Bioinformatics, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Mohammed Yaseen
- Centre for Microvascular Research, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Haitao Wang
- Centre for Microvascular Research, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Laura Vazquez-Martinez
- Centre for Microvascular Research, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Matthew Golding
- Centre for Microvascular Research, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Isobel A Blacksell
- Centre for Biochemical Pharmacology, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Meredith J Giblin
- Centre for Microvascular Research, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Edyta Jaworska
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Cleo L Bishop
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Mathieu-Benoit Voisin
- Centre for Microvascular Research, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Carles Gaston-Massuet
- Centre for Endocrinology, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Liliane Fossati-Jimack
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Costantino Pitzalis
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Dianne Cooper
- Centre for Biochemical Pharmacology, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Thomas D Nightingale
- Centre for Microvascular Research, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Carlos Lopez-Otin
- Centre de Recherche des Cordeliers, Inserm U1138, Université Paris Cité, Sorbonne Université, Paris, France
- Facultad de Ciencias de la Vida y la Naturaleza, Universidad Nebrija, Madrid, Spain
| | - Myles J Lewis
- Centre for Translational Bioinformatics, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Sussan Nourshargh
- Centre for Microvascular Research, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK.
- Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, London, UK.
| |
Collapse
|
42
|
Pan Y, Wang M, Wang P, Wei H, Wei X, Wang D, Hao Y, Wang Y, Chen H. Effects of a semi-interpenetrating network hydrogel loaded with oridonin and DNase-I on the healing of chemoradiotherapy-induced oral mucositis. Biomater Sci 2024; 12:4452-4470. [PMID: 39052032 DOI: 10.1039/d4bm00114a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2024]
Abstract
The aim of this study was to develop a semi-interpenetrating network (IPN) hydrogel system suitable for the oral environment, capable of controlled release of DNase-I and oridonin (ORI), to exert antimicrobial, anti-inflammatory, and reparative effects on chemoradiotherapy-induced oral mucositis (OM). This IPN was based on the combination of ε-polylysine (PLL) and hetastarch (HES), loaded with DNase-I and ORI (ORI/DNase-I/IPN) for OM treatment. In vitro studies were conducted to evaluate degradation, adhesion, release analysis, and bioactivity including cell proliferation and wound healing assays using epidermal keratinocyte and fibroblast cell lines. Furthermore, the therapeutic effects of ORI/DNase-I/IPN were investigated in vivo using Sprague-Dawley (SD) rats with chemoradiotherapy-induced OM. The results demonstrated that the IPN exhibited excellent adhesion to wet mucous membranes, and the two drugs co-encapsulated in the hydrogel were released in a controlled manner, exerting inhibitory effects on bacteria and degrading NETs in wound tissues. The in vivo wound repair effect, microbiological assays, H&E and Masson staining supported the non-toxicity of ORI/DNase-I/IPN, as well as its ability to accelerate the healing of oral ulcers and reduce inflammation. Overall, ORI/DNase-I/IPN demonstrated a therapeutic effect on OM in rats by significantly accelerating the healing process. These findings provide new insights into possible therapies for OM.
Collapse
Affiliation(s)
- Yuxue Pan
- The Key Laboratory of Biomedical Material, School of Life Science and Technology, Xinxiang Medical University, Xinxiang, PR China.
| | - Mengyuan Wang
- The Key Laboratory of Biomedical Material, School of Life Science and Technology, Xinxiang Medical University, Xinxiang, PR China.
| | - Peng Wang
- The Key Laboratory of Biomedical Material, School of Life Science and Technology, Xinxiang Medical University, Xinxiang, PR China.
| | - Hongliang Wei
- School of Chemistry and Chemical Engineering, Henan University of Technology, Zhengzhou, PR China
| | - Xiangjuan Wei
- Clinical Medical Center of Tissue Engineering and Regeneration, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, PR China
| | - Dongmei Wang
- Clinical Medical Center of Tissue Engineering and Regeneration, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, PR China
| | - Yongwei Hao
- The Key Laboratory of Biomedical Material, School of Life Science and Technology, Xinxiang Medical University, Xinxiang, PR China.
| | - Yongxue Wang
- The Key Laboratory of Biomedical Material, School of Life Science and Technology, Xinxiang Medical University, Xinxiang, PR China.
| | - Hongli Chen
- The Key Laboratory of Biomedical Material, School of Life Science and Technology, Xinxiang Medical University, Xinxiang, PR China.
| |
Collapse
|
43
|
Bai L, Wang Y. Mesenchymal stem cells-derived exosomes alleviate senescence of retinal pigment epithelial cells by activating PI3K/AKT-Nrf2 signaling pathway in early diabetic retinopathy. Exp Cell Res 2024; 441:114170. [PMID: 39019426 DOI: 10.1016/j.yexcr.2024.114170] [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: 01/31/2024] [Revised: 06/22/2024] [Accepted: 07/14/2024] [Indexed: 07/19/2024]
Abstract
Diabetic retinopathy (DR) is a major cause of vision loss and blindness in adults. Cellular senescence was involved in the pathogenesis of early-stage DR and is positively correlated with progression. Thus, our study aimed at exploring the effect and potential mechanism of Mesenchymal stem cells-derived exosomes (MSCs-EXOs) on Retinal Pigment Epithelial (RPE) cells senescence at an early stage of DR in vivo and in vitro. ARPE-19 cells were incubated in high glucose (HG) medium mixed with MSCs-EXOs to observe the changes in cell viability. Senescence-associated β-galactosidase (SA-β-gal) staining, Western blot and qRT-PCR were used to assess the expression of senescence-related genes and antioxidant mediators. Quantitative Real-Time polymerase chain reaction (qRT-PCR), Optical coherence tomography (OCT) Hematoxylin and eosin (HE) staining and Electroretinogram (ERG) were respectively used to verify cellular senescence, the structure and function of the retina. Our findings demonstrated that MSCs-EXOs inhibited HG-induced senescence in ARPE-19 cells. Furthermore, MSCs-EXOs reduced HG-induced cell apoptosis and oxidative stress levels while promoting cell proliferation. Mechanistically, HG suppressed PI3K/AKT phosphorylation as well as nuclear factor erythroid 2-related factor 2 (Nrf2) expression along with its downstream target gene expression in ARPE-19 cells. However, MSCs-EXOs reversed these changes by alleviating cellular senescence while enhancing antioxidant activity. In line with our results in vitro, MSCs-EXOs significantly ameliorated hyperglycemia-induced senescence in DR mice by downregulating mRNA expression of P53, P21, P16, and SASP. Additionally, MSCs-EXOs improved the functional and structural integrity of the retina in DR mice. Our study revealed the protective effect of MSCs-EXOs on cellular senescence, offering new insights for the treatment of DR.
Collapse
Affiliation(s)
- Lifang Bai
- Aier Academy of Ophthalmology, Central South University, Changsha, Hunan, PR China
| | - Ying Wang
- Aier Academy of Ophthalmology, Central South University, Changsha, Hunan, PR China; Liaoning Aier Eye Hospital, Shenyang, Liaoning Province, China.
| |
Collapse
|
44
|
Dong X, Song Y, Liu Y, Kou X, Yang T, Shi SX, He K, Li Y, Li Z, Yao X, Guo J, Cui B, Wu Z, Lei Y, Du M, Chen M, Xu H, Liu Q, Shi FD, Wang X, Yan H. Natural killer cells promote neutrophil extracellular traps and restrain macular degeneration in mice. Sci Transl Med 2024; 16:eadi6626. [PMID: 39141700 DOI: 10.1126/scitranslmed.adi6626] [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: 05/10/2023] [Revised: 04/08/2024] [Accepted: 07/23/2024] [Indexed: 08/16/2024]
Abstract
Neovascular age-related macular degeneration (nvAMD) is the leading cause of blindness in the elderly population. Although it is known that nvAMD is associated with focal inflammation, understanding of the precise immune components governing this process remains limited. Here, we identified natural killer (NK) cells as a prominent lymphocyte population infiltrating the perivascular space of choroidal neovascularization (CNV) lesions in patients with nvAMD and in mouse models. Olink proteomic analysis and single-cell RNA sequencing combined with knockout studies demonstrated the involvement of C-C chemokine receptor 5 (CCR5) in NK cell recruitment and extravasation at the CNV sites of mice. Depletion of NK cells or inhibition of activating receptor NK group 2, member D (NKG2D) inhibited the formation of neutrophil extracellular traps, increased vascular leakage, and exacerbated pathological angiogenesis, indicating that NK cells restrain pathogenesis in this mouse model. Age is the strongest risk factor for AMD, and we show that NK cells from aged human donors exhibited a less cytotoxic phenotype. NK cells from old mice exhibited compromised protective effects in the CNV mouse model. In addition, interleukin-2 complex-mediated expansion of NK cells improved CNV formation in mice. Collectively, our study highlights NK cells as a potential therapeutic target for patients with nvAMD.
Collapse
Affiliation(s)
- Xue Dong
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Institute of Eye Health and Eye Diseases, China-UK "Belt and Road" Ophthalmology Joint Laboratory, Tianjin Medical University General Hospital, Tianjin 300052, China
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Yinting Song
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Institute of Eye Health and Eye Diseases, China-UK "Belt and Road" Ophthalmology Joint Laboratory, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Yuming Liu
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Institute of Eye Health and Eye Diseases, China-UK "Belt and Road" Ophthalmology Joint Laboratory, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Xuejing Kou
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Institute of Eye Health and Eye Diseases, China-UK "Belt and Road" Ophthalmology Joint Laboratory, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Tianjing Yang
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Samuel X Shi
- Clinical Neuroscience Research Center (CNRC), Department of Neurosurgery and Neurology, Tulane University School of Medicine, New Orleans, LA 70122, USA
| | - Kai He
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Institute of Eye Health and Eye Diseases, China-UK "Belt and Road" Ophthalmology Joint Laboratory, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Yiming Li
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Institute of Eye Health and Eye Diseases, China-UK "Belt and Road" Ophthalmology Joint Laboratory, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Ziqi Li
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Institute of Eye Health and Eye Diseases, China-UK "Belt and Road" Ophthalmology Joint Laboratory, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Xueming Yao
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Institute of Eye Health and Eye Diseases, China-UK "Belt and Road" Ophthalmology Joint Laboratory, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Ju Guo
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Institute of Eye Health and Eye Diseases, China-UK "Belt and Road" Ophthalmology Joint Laboratory, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Bohao Cui
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Institute of Eye Health and Eye Diseases, China-UK "Belt and Road" Ophthalmology Joint Laboratory, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Ziru Wu
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Yi Lei
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Institute of Eye Health and Eye Diseases, China-UK "Belt and Road" Ophthalmology Joint Laboratory, Tianjin Medical University General Hospital, Tianjin 300052, China
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Mei Du
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Institute of Eye Health and Eye Diseases, China-UK "Belt and Road" Ophthalmology Joint Laboratory, Tianjin Medical University General Hospital, Tianjin 300052, China
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Mei Chen
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Heping Xu
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Qiang Liu
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Fu-Dong Shi
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Xiaohong Wang
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Institute of Eye Health and Eye Diseases, China-UK "Belt and Road" Ophthalmology Joint Laboratory, Tianjin Medical University General Hospital, Tianjin 300052, China
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Hua Yan
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Institute of Eye Health and Eye Diseases, China-UK "Belt and Road" Ophthalmology Joint Laboratory, Tianjin Medical University General Hospital, Tianjin 300052, China
- School of Medicine, Nankai University, Tianjin 300071, China
| |
Collapse
|
45
|
Bergersen KV, Kavvathas B, Ford BD, Wilson EH. Toxoplasma infection induces an aged neutrophil population in the CNS that is associated with neuronal protection. J Neuroinflammation 2024; 21:189. [PMID: 39095837 PMCID: PMC11297776 DOI: 10.1186/s12974-024-03176-7] [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: 05/22/2024] [Accepted: 07/15/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND Infection with the protozoan parasite Toxoplasma gondii leads to the formation of lifelong cysts in neurons that can have devastating consequences in the immunocompromised. In the immunocompetent individual, anti-parasitic effector mechanisms and a balanced immune response characterized by pro- and anti-inflammatory cytokine production establishes an asymptomatic infection that rarely leads to neurological symptoms. Several mechanisms are known to play a role in this successful immune response in the brain including T cell production of IFNγ and IL-10 and the involvement of CNS resident cells. This limitation of clinical neuropathology during chronic infection suggests a balance between immune response and neuroprotective mechanisms that collectively prevent clinical manifestations of disease. However, how these two vital mechanisms of protection interact during chronic Toxoplasma infection remains poorly understood. MAIN TEXT This study demonstrates a previously undescribed connection between innate neutrophils found chronically in the brain, termed "chronic brain neutrophils" (CBNeuts), and neuroprotective mechanisms during Toxoplasma infection. Lack of CBNeuts during chronic infection, accomplished via systemic neutrophil depletion, led to enhanced infection and deleterious effects on neuronal regeneration and repair mechanisms in the brain. Phenotypic and transcriptomic analysis of CBNeuts identified them as distinct from peripheral neutrophils and revealed two main subsets of CBNeuts that display heterogeneity towards both classical effector and neuroprotective functions in an age-dependent manner. Further phenotypic profiling defined expression of the neuroprotective molecules NRG-1 andErbB4 by these cells, and the importance of this signaling pathway during chronic infection was demonstrated via NRG-1 treatment studies. CONCLUSIONS In conclusion, this work identifies CBNeuts as a heterogenous population geared towards both classical immune responses and neuroprotection during chronic Toxoplasma infection and provides the foundation for future mechanistic studies of these cells.
Collapse
Affiliation(s)
- Kristina V Bergersen
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, USA
| | - Bill Kavvathas
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, USA
| | - Byron D Ford
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, USA
- College of Medicine, Howard University, Washington, D.C., USA
| | - Emma H Wilson
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, USA.
| |
Collapse
|
46
|
Terao R, Sohn BS, Yamamoto T, Lee TJ, Colasanti J, Pfeifer CW, Lin JB, Santeford A, Yamaguchi S, Yoshida M, Apte RS. Cholesterol Accumulation Promotes Photoreceptor Senescence and Retinal Degeneration. Invest Ophthalmol Vis Sci 2024; 65:29. [PMID: 39167399 PMCID: PMC11343002 DOI: 10.1167/iovs.65.10.29] [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: 04/28/2024] [Accepted: 08/01/2024] [Indexed: 08/23/2024] Open
Abstract
Purpose Dysregulated cholesterol metabolism is critical in the pathogenesis of AMD. Cellular senescence contributes to the development of numerous age-associated diseases. In this study, we investigated the link between cholesterol burden and the cellular senescence of photoreceptors. Methods Retinas from rod-specific ATP binding cassette subfamily A member 1 (Abca1) and G member 1 (Abcg1) (Abca1/g1-rod/-rod) knockout mice fed with a high-fat diet were analyzed for the signs of cellular senescence. Real-time quantitative PCR and immunofluorescence were used to characterize the senescence profile of the retina and cholesterol-treated photoreceptor cell line (661W). Inducible elimination of p16(Ink4a)-positive senescent cells (INK-ATTAC) mice or the administration of senolytic drugs (dasatinib and quercetin: D&Q) were used to examine the impact of senolytics on AMD-like phenotypes in Abca1/g1-rod/-rod retina. Results Increased accumulation of senescent cells as measured by markers of cellular senescence was found in Abca1/g1-rod/-rod retina. Exogenous cholesterol also induced cellular senescence in 661W cells. Selective elimination of senescent cells in Abca1/g1-rod/-rod;INK-ATTAC mice or by administration of D&Q improved visual function, lipid accumulation in retinal pigment epithelium, and Bruch's membrane thickening. Conclusions Cholesterol accumulation promotes cellular senescence in photoreceptors. Eliminating senescent photoreceptors improves visual function in a model of retinal neurodegeneration, and senotherapy offers a novel therapeutic avenue for further investigation.
Collapse
Affiliation(s)
- Ryo Terao
- John F. Hardesty, MD, Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, St. Louis, Missouri, United States
- Department of Ophthalmology, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
| | - Brian S. Sohn
- John F. Hardesty, MD, Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Taku Yamamoto
- John F. Hardesty, MD, Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, St. Louis, Missouri, United States
- Department of Ophthalmology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Hokkaido, Japan
| | - Tae Jun Lee
- John F. Hardesty, MD, Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Jason Colasanti
- John F. Hardesty, MD, Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Charles W. Pfeifer
- John F. Hardesty, MD, Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Joseph B. Lin
- John F. Hardesty, MD, Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Andrea Santeford
- John F. Hardesty, MD, Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Shinobu Yamaguchi
- John F. Hardesty, MD, Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Mitsukuni Yoshida
- John F. Hardesty, MD, Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, St. Louis, Missouri, United States
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Rajendra S. Apte
- John F. Hardesty, MD, Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, St. Louis, Missouri, United States
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, United States
| |
Collapse
|
47
|
Chesnokova V, Zonis S, Apaydin T, Barrett R, Melmed S. Non-pituitary growth hormone enables colon cell senescence evasion. Aging Cell 2024; 23:e14193. [PMID: 38724466 PMCID: PMC11320355 DOI: 10.1111/acel.14193] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 03/18/2024] [Accepted: 04/02/2024] [Indexed: 08/15/2024] Open
Abstract
DNA damage-induced senescence is initially sustained by p53. Senescent cells produce a senescence-associated secretory phenotype (SASP) that impacts the aging microenvironment, often promoting cell transformation. Employing normal non-tumorous human colon cells (hNCC) derived from surgical biopsies and three-dimensional human intestinal organoids, we show that local non-pituitary growth hormone (npGH) induced in senescent cells is a SASP component acting to suppress p53. npGH autocrine/paracrine suppression of p53 results in senescence evasion and cell-cycle reentry, as evidenced by increased Ki67 and BrdU incorporation. Post-senescent cells exhibit activated epithelial-to-mesenchymal transition (EMT), and increased cell motility. Nu/J mice harboring GH-secreting HCT116 xenografts with resultant high GH levels and injected intrasplenic with post-senescent hNCC developed fourfold more metastases than did mice harboring control xenografts, suggesting that paracrine npGH enables post-senescent cell transformation. By contrast, senescent cells with suppressed npGH exhibit downregulated Ki67 and decreased soft agar colony formation. Mechanisms underlying these observations include npGH induction by the SASP chemokine CXCL1, which attracts immune effectors to eliminate senescent cells; GH, in turn, suppresses CXCL1, likely by inhibiting phospho-NFκB, resulting in SASP cytokine downregulation. Consistent with these findings, GH-receptor knockout mice exhibited increased colon phospho-NFκB and CXCL1, while GH excess decreased colon CXCL1. The results elucidate mechanisms for local hormonal regulation of microenvironmental changes in DNA-damaged non-tumorous epithelial cells and portray a heretofore unappreciated GH action favoring age-associated epithelial cell transformation.
Collapse
Affiliation(s)
- Vera Chesnokova
- Department of MedicineCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | - Svetlana Zonis
- Department of MedicineCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | - Tugce Apaydin
- Department of MedicineCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | - Robert Barrett
- Board of Governors Regenerative Medicine InstituteCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | - Shlomo Melmed
- Department of MedicineCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| |
Collapse
|
48
|
Wang X, Wang T, Kaneko S, Kriukov E, Lam E, Szczepan M, Chen J, Gregg A, Wang X, Fernandez-Gonzalez A, Mitsialis SA, Kourembanas S, Baranov P, Sun Y. Photoreceptors inhibit pathological retinal angiogenesis through transcriptional regulation of Adam17 via c-Fos. Angiogenesis 2024; 27:379-395. [PMID: 38483712 PMCID: PMC11303108 DOI: 10.1007/s10456-024-09912-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 03/03/2024] [Indexed: 04/11/2024]
Abstract
Pathological retinal angiogenesis profoundly impacts visual function in vascular eye diseases, such as retinopathy of prematurity (ROP) in preterm infants and age-related macular degeneration in the elderly. While the involvement of photoreceptors in these diseases is recognized, the underlying mechanisms remain unclear. This study delved into the pivotal role of photoreceptors in regulating abnormal retinal blood vessel growth using an oxygen-induced retinopathy (OIR) mouse model through the c-Fos/A disintegrin and metalloprotease 17 (Adam17) axis. Our findings revealed a significant induction of c-Fos expression in rod photoreceptors, and c-Fos depletion in these cells inhibited pathological neovascularization and reduced blood vessel leakage in the OIR mouse model. Mechanistically, c-Fos directly regulated the transcription of Adam17 a shedding protease responsible for the production of bioactive molecules involved in inflammation, angiogenesis, and cell adhesion and migration. Furthermore, we demonstrated the therapeutic potential by using an adeno-associated virus carrying a rod photoreceptor-specific short hairpin RNA against c-fos which effectively mitigated abnormal retinal blood vessel overgrowth, restored retinal thickness, and improved electroretinographic (ERG) responses. In conclusion, this study highlights the significance of photoreceptor c-Fos in ROP pathology, offering a novel perspective for the treatment of this disease.
Collapse
Affiliation(s)
- Xudong Wang
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Tianxi Wang
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Satoshi Kaneko
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Emil Kriukov
- Department of Ophthalmology, The Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Enton Lam
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Manon Szczepan
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jasmine Chen
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Austin Gregg
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Xingyan Wang
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Angeles Fernandez-Gonzalez
- Division of Newborn Medicine, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - S Alex Mitsialis
- Division of Newborn Medicine, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Stella Kourembanas
- Division of Newborn Medicine, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Petr Baranov
- Department of Ophthalmology, The Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Ye Sun
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
49
|
Li Y, Hu B, Lu L, Li Y, Caika S, Song Z, Sen G. Development and external validation of a predictive model for type 2 diabetic retinopathy. Sci Rep 2024; 14:16741. [PMID: 39033211 PMCID: PMC11271465 DOI: 10.1038/s41598-024-67533-5] [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: 12/15/2023] [Accepted: 07/12/2024] [Indexed: 07/23/2024] Open
Abstract
Diabetes retinopathy (DR) is a critical clinical disease with that causes irreversible visual damage in adults, and may even lead to permanent blindness in serious cases. Early identification and treatment of DR is critical. Our aim was to train and externally validate a prediction nomogram for early prediction of DR. 2381 patients with type 2 diabetes mellitus (T2DM) were retrospective study from the First Affiliated Hospital of Xinjiang Medical University in Xinjiang, China, hospitalised between Jan 1, 2019 and Jun 30, 2022. 962 patients with T2DM from the Suzhou BenQ Hospital in Jiangsu, China hospitalised between Jul 1, 2020 to Jun 30, 2022 were considered for external validation. The least absolute shrinkage and selection operator (LASSO) and multivariate logistic regression was performed to identify independent predictors and establish a nomogram to predict the occurrence of DR. The performance of the nomogram was evaluated using a receiver operating characteristic curve (ROC), a calibration curve, and decision curve analysis (DCA). Neutrophil, 25-hydroxyvitamin D3 [25(OH)D3], Duration of T2DM, hemoglobin A1c (HbA1c), and Apolipoprotein A1 (ApoA1) were used to establish a nomogram model for predicting the risk of DR. In the development and external validation groups, the areas under the curve of the nomogram constructed from the above five factors were 0.834 (95%CI 0.820-0.849) and 0.851 (95%CI 0.829-0.874), respectively. The nomogram demonstrated excellent performance in the calibration curve and DCA. This research has developed and externally verified that the nomograph model shows a good predictive ability in assessing DR risk in people with type 2 diabetes. The application of this model will help clinicians to intervene early, thus effectively reducing the incidence rate and mortality of DR in the future, and has far-reaching significance in improving the long-term health prognosis of diabetes patients.
Collapse
Affiliation(s)
- Yongsheng Li
- Department of Preventive Medicine, Medical College, Tarim University, Alar, 843300, China
| | - Bin Hu
- Department of Preventive Medicine, Medical College, Tarim University, Alar, 843300, China
| | - Lian Lu
- Department of Medical Engineering and Technology, Xinjiang Medical University, Ürümqi, 830011, China
| | - Yongnan Li
- Nursing Department, Suzhou BenQ Hospital, Suzhou, 215163, China
| | - Siqingaowa Caika
- Nursing Department, First Affiliated Hospital of Xinjiang Medical University, Ürümqi, 830054, China
| | - Zhixin Song
- Department of Medical Engineering and Technology, Xinjiang Medical University, Ürümqi, 830011, China
| | - Gan Sen
- Department of Medical Engineering and Technology, Xinjiang Medical University, Ürümqi, 830011, China.
| |
Collapse
|
50
|
Bammidi S, Koontz V, Gautam P, Hose S, Sinha D, Ghosh S. Neutrophils in Ocular Diseases. Int J Mol Sci 2024; 25:7736. [PMID: 39062975 PMCID: PMC11276787 DOI: 10.3390/ijms25147736] [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: 04/22/2024] [Revised: 07/09/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
Neutrophils, traditionally viewed as first responders to infection or tissue damage, exhibit dynamic and diverse roles in ocular health and disease. This review elaborates on previous findings that showed how neutrophils contribute to ocular diseases. In ocular infections, neutrophils play a pivotal role in host defense by orchestrating inflammatory responses to combat pathogens. Furthermore, in optic nerve neuropathies and retinal degenerative diseases like age-related macular degeneration (AMD) and diabetic retinopathy (DR), neutrophils are implicated in neuroinflammation and tissue damage owing to their ability to undergo neutrophil extracellular trap formation (NETosis) and secretion of inflammatory molecules. Targeting neutrophil-dependent processes holds promise as a therapeutic strategy, offering potential avenues for intervention in ocular infections, cancers, and retinal degenerative diseases. Understanding the multifaceted roles of neutrophils in ocular diseases is crucial for developing targeted therapies to improve patient outcomes.
Collapse
Affiliation(s)
- Sridhar Bammidi
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA; (S.B.); (V.K.); (P.G.); (S.H.); (D.S.)
| | - Victoria Koontz
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA; (S.B.); (V.K.); (P.G.); (S.H.); (D.S.)
| | - Pooja Gautam
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA; (S.B.); (V.K.); (P.G.); (S.H.); (D.S.)
| | - Stacey Hose
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA; (S.B.); (V.K.); (P.G.); (S.H.); (D.S.)
| | - Debasish Sinha
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA; (S.B.); (V.K.); (P.G.); (S.H.); (D.S.)
- The Wilmer Eye Institute, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Sayan Ghosh
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA; (S.B.); (V.K.); (P.G.); (S.H.); (D.S.)
| |
Collapse
|