1
|
Zhu T, Ding Y, Wu X, Li Y, Cheng G, Wang N, Yang Q, Zhang W, Chen X, Liu X. Pentraxin 3 promotes the expression of pro-inflammatory cytokines and the migration of macrophages in myocarditis. BMC Cardiovasc Disord 2025; 25:354. [PMID: 40335910 PMCID: PMC12060373 DOI: 10.1186/s12872-025-04790-w] [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: 11/24/2024] [Accepted: 04/21/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND This study aims to investigate the expression of Pentraxin 3 (PTX3) and Nod-like receptor family pyrin domain-containing 3 (NLRP3) in myocarditis and to elucidate their roles and potential interplay in the pathogenesis of myocarditis. METHODS Immunofluorescence staining was performed on myocardial autopsy specimens from deceased patients with severe myocarditis or severe trauma. H9C2 cardiomyocytes were divided into five groups: Control, Lipopolysaccharide (LPS), LPS + PTX3 overexpression, LPS + small interfering RNA targeting PTX3 (si-PTX3), and LPS + PTX3 overexpression + si-NLRP3. The expression levels of PTX3 and NLRP3 at the RNA level were quantified using quantitative real-time polymerase chain reaction (qPCR), while protein expression was assessed via western blot. The concentrations of interleukin-1β (IL-1β) and IL-18 were determined by enzyme-linked immunosorbent assay (ELISA). Macrophages migration was evaluated using Transwell assays. RESULTS Immunofluorescence staining revealed co-localization and increased expression of PTX3 and NLRP3 in the myocardium of patients with severe myocarditis. In vitro experiments demonstrated that PTX3 enhanced the expression of NLRP3, IL-1β, and IL-18 in LPS-stimulated cardiomyocytes. Furthermore, PTX3 was shown to promote macrophage migration by regulating NLRP3 expression, as assessed by Transwell assays. CONCLUSION Our findings suggest that PTX3-mediated NLRP3 activation contributes to inflammatory responses and promotes macrophage migration in myocarditis. This study provides a foundation for future investigations into PTX3-targeted therapies for myocarditis.
Collapse
Affiliation(s)
- Tianyu Zhu
- Department of Cardiology, Peking University International Hospital, Beijing, 102206, P.R. China
| | - Ying Ding
- Department of Nephrology, The Second Medical Center of Chinese PLA General Hospital, National Clinical Research Centre for Geriatric Diseases, Beijing, 100853, P.R. China
| | - Xiaohui Wu
- Department of Cardiology, Peking University International Hospital, Beijing, 102206, P.R. China
| | - Yan Li
- Department of Cardiology, Peking University International Hospital, Beijing, 102206, P.R. China
| | - Guanliang Cheng
- Department of Cardiology, Peking University International Hospital, Beijing, 102206, P.R. China
| | - Ning Wang
- Department of Cardiology, Peking University International Hospital, Beijing, 102206, P.R. China
| | - Quan Yang
- Department of Cardiology, Peking University International Hospital, Beijing, 102206, P.R. China
| | - Wenchao Zhang
- Department of Cardiology, Peking University International Hospital, Beijing, 102206, P.R. China
| | - Xuezhi Chen
- Department of Cardiology, Peking University International Hospital, Beijing, 102206, P.R. China.
| | - Xiaohui Liu
- Department of Cardiology, Peking University International Hospital, Beijing, 102206, P.R. China
| |
Collapse
|
2
|
Jiang M, Cai N, Hu J, Han L, Xu F, Zhu B, Wang B. Genomic and algorithm-based predictive risk assessment models for benzene exposure. Front Public Health 2025; 12:1419361. [PMID: 39911783 PMCID: PMC11795664 DOI: 10.3389/fpubh.2024.1419361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 12/12/2024] [Indexed: 02/07/2025] Open
Abstract
Aim In this research, we leveraged bioinformatics and machine learning to pinpoint key risk genes associated with occupational benzene exposure and to construct genomic and algorithm-based predictive risk assessment models. Subject and methods We sourced GSE9569 and GSE21862 microarray data from the Gene Expression Omnibus. Utilizing R software, we performed an initial screen for differentially expressed genes (DEGs), which was followed by the enrichment analyses to elucidate the affected functions and pathways. Subsequent steps included the application of three machine learning algorithms for key gene identification, and the validation of these genes within both a cohort exposed to benzene and a benzene-exposed mice model. We then conducted a functional prediction analysis on these genes using four machine learning models, complemented by GSVA enrichment analysis. Results Out of the data, 40 DEGs were identified, primarily linked to cytokine signaling, lipopolysaccharide response, and chemokine pathways. NFKB1, PHACTR1, PTGS2, and PTX3 were pinpointed as significant through machine learning. Validation confirmed substantial changes in NFKB1 and PTX3 following exposure, with PTX3 emerging as paramount, suggesting its utility as a diagnostic biomarker for benzene damage. Conclusion Risk assessment models, informed by oxidative stress markers, successfully discriminated between benzene-injured patients and controls.
Collapse
Affiliation(s)
- Minyun Jiang
- School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Province Center for Disease Prevention and Control, Institute of Occupational Disease Prevention, Nanjing, Jiangsu, China
| | - Na Cai
- School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Province Center for Disease Prevention and Control, Institute of Occupational Disease Prevention, Nanjing, Jiangsu, China
| | - Juan Hu
- School of Public Health, Southeast University, Nanjing, Jiangsu, China
| | - Lei Han
- Jiangsu Province Center for Disease Prevention and Control, Institute of Occupational Disease Prevention, Nanjing, Jiangsu, China
- Jiangsu Preventive Medical Association, Nanjing, Jiangsu, China
| | - Fanwei Xu
- School of Public Health, Southeast University, Nanjing, Jiangsu, China
| | - Baoli Zhu
- School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Province Center for Disease Prevention and Control, Institute of Occupational Disease Prevention, Nanjing, Jiangsu, China
- Jiangsu Preventive Medical Association, Nanjing, Jiangsu, China
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Boshen Wang
- Jiangsu Province Center for Disease Prevention and Control, Institute of Occupational Disease Prevention, Nanjing, Jiangsu, China
| |
Collapse
|
3
|
Das S, Ramanathan G. Assessing the Inhibitory Potential of Pregnenolone Sulfate on Pentraxin 3 in Diabetic Kidney Disease: A Molecular Docking and Simulation Study. J Cell Biochem 2025; 126:e30661. [PMID: 39344977 DOI: 10.1002/jcb.30661] [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: 07/09/2024] [Revised: 09/07/2024] [Accepted: 09/11/2024] [Indexed: 10/01/2024]
Abstract
Diabetic Kidney Disease (DKD), a frequent consequence of diabetes, has substantial implications for both morbidity and mortality rates, prompting the exploration of new metabolic biomarkers due to limitations in current methods like creatinine and albumin measurements. Pentraxin 3 (PTX3) shows promise for assessing renal inflammation in DKD. This study investigates how DKD metabolites could influence PTX3 expression through molecular docking, ADMET profiling, and dynamic simulation. Network and pathway analyses were conducted to explore metabolite interactions with DKD genes and their contributions to DKD pathogenesis. Thirty-three DKD-associated metabolites were screened, using pentoxifylline (PEN) as a reference. The pharmacokinetic properties of these compounds were evaluated through molecular docking and ADMET profiling. Molecular dynamics simulations over 200 ns assessed the stability of PTX3 (apo), the PRE-PTX3 complex, and PEN-PTX3 across multiple parameters. Cytoscape identified 1082 nodes and 1381 edges linking metabolites with DKD genes. KEGG pathway analysis underscored PTX3's role in inflammation. Molecular docking revealed pregnenolone sulfate (PRE) with the highest binding affinity (-6.25 kcal/mol), followed by hydrocortisone (-6.03 kcal/mol) and 2-arachidonoylglycerol (-5.92 kcal/mol), compared to PEN (-5.35 kcal/mol). ADMET profiling selected PRE for dynamic simulation alongside PEN. Analysis of RMSD, RMSF, RG, SASA, H-bond, PCA, FEL, and MM-PBSA indicated stable complex behavior over time. Our findings suggest that increasing PRE levels could be beneficial in managing DKD, potentially through isolating PRE from fungal sources, synthesizing it as dietary supplements, or enhancing endogenous PRE synthesis within the body.
Collapse
Affiliation(s)
- Soumik Das
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India
| | - Gnanasambandan Ramanathan
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India
| |
Collapse
|
4
|
Mulat SY, Mihajlović M, Antonić T, Miloševski-Lomić G, Peco-Antić A, Jovanović D, Paripović D, Stefanović A. Pediatric nephrotic syndrome: The interplay of oxidative stress and inflammation. J Med Biochem 2024; 43:424-435. [PMID: 39139165 PMCID: PMC11318042 DOI: 10.5937/jomb0-46526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 12/18/2023] [Indexed: 08/15/2024] Open
Abstract
Background The pathophysiological mechanisms crucial in the development of nephrotic syndrome (NS) in the pediatric population are still not fully understood. This study aimed to investigate the relationship between hypertension, oxidative stress, and inflammation in pediatric patients during the acute phase of the disease. Methods The study included 33 children, aged 2 to 9 years, with nephrotic syndrome. Blood samples were collected during the acute phase and remission. Parameters of oxidative status were determined, including total oxidative status (TOS), advanced oxidation protein products (AOPP), prooxidant-antioxidant balance (PAB), sulfhydryl groups (-SH), paraoxonase 1 (PON1), and total antioxidant status (TAS) in serum, measured spectrophotometrically. Inflam - matory parameters such as pentraxin 3 (PTX3), leptin, program med cell death ligand 1 (PD-L1), and E-cadherin were determined using enzyme-linked immunosorbent assay (ELISA). Results Patients with nephrotic syndrome and hypertension had significantly higher levels of advanced oxidation protein products and total antioxidant status (p=0.029 and p=0.003, respectively). During the acute phase of the disease, lower activity of sulfhydryl groups and paraoxonase 1 was observed compared to remission (p<0.001, for both). Pentraxin 3 levels were higher, while leptin levels were lower during the acute phase (p<0.001, for both). Pentraxin 3 correlated with advanced oxidation protein products and total antioxidant status during the acute phase but not in remission (rs=0.42, p=0.027 and rs=0.43, p=0.025, respectively). A negative correlation between Advanced oxidation protein products and leptin was observed during the acute phase, which disappeared in remission (rs=-0.42, p=0.028). Conclusions Results of this study show that hypertension influences oxidative stress markers, and decreased antioxidant capacity may contribute to nephrotic syndrome development. Pentraxin 3 appears as a potential disease activity marker, indicating a dynamic connection between inflammation and oxidative stress. Leptin may also play a role in oxidative stress in nephrotic syndrome.
Collapse
Affiliation(s)
- Simachew Yonas Mulat
- University of Belgrade, Faculty of Pharmacy, Department of Medical Biochemistry, Belgrade
| | - Marija Mihajlović
- University of Belgrade, Faculty of Pharmacy, Department of Medical Biochemistry, Belgrade
| | - Tamara Antonić
- University of Belgrade, Faculty of Pharmacy, Department of Medical Biochemistry, Belgrade
| | | | - Amira Peco-Antić
- University Children's Hospital, Department of Nephrology, Belgrade
| | | | - Dušan Paripović
- University Children's Hospital, Department of Nephrology, Belgrade
| | - Aleksandra Stefanović
- University of Belgrade, Faculty of Pharmacy, Department of Medical Biochemistry, Belgrade
| |
Collapse
|
5
|
Santos-Ribeiro D, Cunha C, Carvalho A. Humoral pathways of innate immune regulation in granuloma formation. Trends Immunol 2024; 45:419-427. [PMID: 38762333 DOI: 10.1016/j.it.2024.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 04/24/2024] [Accepted: 04/24/2024] [Indexed: 05/20/2024]
Abstract
The humoral arm of mammalian innate immunity regulates several molecular mechanisms involved in resistance to pathogens, inflammation, and tissue repair. Recent studies highlight the crucial role played by humoral mediators in granulomatous inflammation. However the molecular mechanisms linking the function of these soluble molecules to the initiation and maintenance of granulomas remain elusive. We propose that humoral innate immunity coordinates fundamental physiological processes in macrophages which, in turn, initiate activation and transformation events that enable granuloma formation. We discuss the involvement of humoral mediators in processes such as immune activation, phagocytosis, metabolism, and tissue remodeling, and how these can dictate macrophage functionality during granuloma formation. These advances present opportunities for discovering novel disease factors and developing targeted, more effective treatments for granulomatous diseases.
Collapse
Affiliation(s)
- Diana Santos-Ribeiro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Cristina Cunha
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Agostinho Carvalho
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
6
|
Ye X, Wang Z, Lei W, Shen M, Tang J, Xu X, Yang Y, Zhang H. Pentraxin 3: A promising therapeutic target for cardiovascular diseases. Ageing Res Rev 2024; 93:102163. [PMID: 38092307 DOI: 10.1016/j.arr.2023.102163] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 11/23/2023] [Accepted: 12/07/2023] [Indexed: 12/18/2023]
Abstract
Cardiovascular disease (CVD) is the primary global cause of death, and inflammation is a crucial factor in the development of CVDs. The acute phase inflammatory protein pentraxin 3 (PTX3) is a biomarker reflecting the immune response. Recent research indicates that PTX3 plays a vital role in CVDs and has been investigated as a possible biomarker for CVD in clinical trials. PTX3 is implicated in the progression of CVDs through mechanisms such as exacerbating vascular endothelial dysfunction, affecting angiogenesis, and regulating inflammation and oxidative stress. This review summarized the structure and function of PTX3, focusing on its multifaceted effects on CVDs, such as atherosclerosis, myocardial infarction, and hypertension. This may help in explaining the varying PTX3 functions and usage, as well as in utilizing target organs to manage diseases. Moreover, elucidating the opposite role of PTX3 in the cardiovascular system will demonstrate the therapeutic and predictive potential in human diseases.
Collapse
Affiliation(s)
- Xingyan Ye
- Department of Cardiology, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University. Faculty of Life Sciences and Medicine, Northwest University, 10 Fengcheng Three Road, Xi'an, China; Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, China
| | - Zheng Wang
- Department of Cardiothoracic Surgery, Central Theater Command General Hospital of Chinese People's Liberation Army, 627 Wuluo Road, Wuhan, China
| | - Wangrui Lei
- Department of Cardiology, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University. Faculty of Life Sciences and Medicine, Northwest University, 10 Fengcheng Three Road, Xi'an, China
| | - Mingzhi Shen
- Department of General Medicine, Hainan Hospital of Chinese People's Liberation Army (PLA) General Hospital, 80 Jianglin Road, Hainan, China
| | - Jiayou Tang
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an, China
| | - Xuezeng Xu
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an, China
| | - Yang Yang
- Department of Cardiology, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University. Faculty of Life Sciences and Medicine, Northwest University, 10 Fengcheng Three Road, Xi'an, China; Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, China.
| | - Huan Zhang
- Department of Cardiology, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University. Faculty of Life Sciences and Medicine, Northwest University, 10 Fengcheng Three Road, Xi'an, China; Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, China.
| |
Collapse
|
7
|
Park HR, Harris SM, Boldenow E, Aronoff DM, Rea M, Xi C, Loch-Caruso R. The antioxidant N-acetyl cysteine inhibits cytokine and prostaglandin release in human fetal membranes stimulated ex vivo with lipoteichoic acid or live group B streptococcus. Am J Reprod Immunol 2024; 91:e13807. [PMID: 38282602 PMCID: PMC10832889 DOI: 10.1111/aji.13807] [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: 09/08/2023] [Revised: 12/15/2023] [Accepted: 12/20/2023] [Indexed: 01/30/2024] Open
Abstract
BACKGROUNDS Infection during pregnancy is a significant public health concern due to the increased risk of adverse birth outcomes. Group B Streptococcus or Streptococcus agalactiae (GBS) stands out as a major bacterial cause of neonatal morbidity and mortality. We aimed to explore the involvement of reactive oxygen species (ROS) and oxidative stress pathways in pro-inflammatory responses within human fetal membrane tissue, the target tissue of acute bacterial chorioamnionitis. METHODS We reanalyzed transcriptomic data from fetal membrane explants inoculated with GBS to assess the impact of GBS on oxidative stress and ROS genes/pathways. We conducted pathway enrichment analysis of transcriptomic data using the Database for Annotation, Visualization and Integrated Discovery (DAVID), a web-based functional annotation/pathway enrichment tool. Subsequently, we conducted ex vivo experiments to test the hypothesis that antioxidant treatment could inhibit pathogen-stimulated inflammatory responses in fetal membranes. RESULTS Using DAVID analysis, we found significant enrichment of pathways related to oxidative stress or ROS in GBS-inoculated human fetal membranes, for example, "Response to Oxidative Stress" (FDR = 0.02) and "Positive Regulation of Reactive Oxygen Species Metabolic Process" (FDR = 2.6*10-4 ). There were 31 significantly changed genes associated with these pathways, most of which were upregulated after GBS inoculation. In ex vivo experiments with choriodecidual membrane explants, our study showed that co-treatment with N-acetylcysteine (NAC) effectively suppressed the release of pro-inflammatory cytokines (IL-6, IL-8, TNF-α) and prostaglandin PGE2, compared to GBS-treated explants (p < .05 compared to GBS-treated samples without NAC co-treatment). Furthermore, NAC treatment inhibited the release of cytokines and PGE2 stimulated by lipoteichoic acid (LTA) and lipopolysaccharide (LPS) in whole membrane explants (p < .05 compared to LTA or LPS-treated samples without NAC co-treatment). CONCLUSIONS Our study sheds light on the potential roles of ROS in governing the innate immune response to GBS infection, offering insights for developing strategies to mitigate GBS-related adverse outcomes.
Collapse
Affiliation(s)
- Hae-Ryung Park
- Department of Environmental Medicine, School of Medicine and Dentistry, University of Rochester, Rochester, NY 14642 USA
| | - Sean M. Harris
- Department of Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, MI 48109-2029 USA
| | - Erica Boldenow
- Department of Biology, Calvin University, Grand Rapids, MI 49546-4402 USA
| | - David M. Aronoff
- Department of Medicine, School of Medicine, Indiana University, Indianapolis, IN 46202-3082
| | - Meaghan Rea
- Department of Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, MI 48109-2029 USA
| | - Chuanwu Xi
- Department of Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, MI 48109-2029 USA
| | - Rita Loch-Caruso
- Department of Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, MI 48109-2029 USA
| |
Collapse
|
8
|
Różanowska MB. Lipofuscin, Its Origin, Properties, and Contribution to Retinal Fluorescence as a Potential Biomarker of Oxidative Damage to the Retina. Antioxidants (Basel) 2023; 12:2111. [PMID: 38136230 PMCID: PMC10740933 DOI: 10.3390/antiox12122111] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 12/05/2023] [Accepted: 12/09/2023] [Indexed: 12/24/2023] Open
Abstract
Lipofuscin accumulates with age as intracellular fluorescent granules originating from incomplete lysosomal digestion of phagocytosed and autophagocytosed material. The purpose of this review is to provide an update on the current understanding of the role of oxidative stress and/or lysosomal dysfunction in lipofuscin accumulation and its consequences, particularly for retinal pigment epithelium (RPE). Next, the fluorescence of lipofuscin, spectral changes induced by oxidation, and its contribution to retinal fluorescence are discussed. This is followed by reviewing recent developments in fluorescence imaging of the retina and the current evidence on the prognostic value of retinal fluorescence for the progression of age-related macular degeneration (AMD), the major blinding disease affecting elderly people in developed countries. The evidence of lipofuscin oxidation in vivo and the evidence of increased oxidative damage in AMD retina ex vivo lead to the conclusion that imaging of spectral characteristics of lipofuscin fluorescence may serve as a useful biomarker of oxidative damage, which can be helpful in assessing the efficacy of potential antioxidant therapies in retinal degenerations associated with accumulation of lipofuscin and increased oxidative stress. Finally, amendments to currently used fluorescence imaging instruments are suggested to be more sensitive and specific for imaging spectral characteristics of lipofuscin fluorescence.
Collapse
Affiliation(s)
- Małgorzata B. Różanowska
- School of Optometry and Vision Sciences, College of Biomedical and Life Sciences, Cardiff University, Maindy Road, Cardiff CF24 4HQ, Wales, UK;
- Cardiff Institute for Tissue Engineering and Repair (CITER), Redwood Building, King Edward VII Avenue, Cardiff CF10 3NB, Wales, UK
| |
Collapse
|
9
|
Bousquet E, Chenevier-Gobeaux C, Jaworski T, Torres-Villaros H, Zola M, Mantel I, Kowalczuk L, Matet A, Daruich A, Zhao M, Yzer S, Behar-Cohen F. High Levels of C-Reactive Protein with Low Levels of Pentraxin 3 as Biomarkers for Central Serous Chorioretinopathy. OPHTHALMOLOGY SCIENCE 2023; 3:100278. [PMID: 36950301 PMCID: PMC10025279 DOI: 10.1016/j.xops.2023.100278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 01/26/2023] [Accepted: 01/27/2023] [Indexed: 02/05/2023]
Abstract
Purpose To investigate the association between the 2 acute phase proteins, C-reactive protein (CRP) and pentraxin 3 (PTX3) with central serous chorioretinopathy (CSCR), as PTX3 is a glucocorticoid-induced protein. Design Cross-sectional multicenter study. Participants Patients with CSCR compared with age- and sex-matched healthy participants. Methods Patients with CSCR from 3 centers in Europe were included in the study. The clinical form of CSCR was recorded. Blood samples from patients with CSCR and healthy participants were sampled, and high-sensitivity CRP and PTX3 levels were measured in the serum. Main Outcome Measures C-reactive protein and PTX3 serum level comparison between patients with CSCR with age- and sex-matched healthy participants. Results Although CRP levels were higher in patients with CSCR (n = 216) than in age- and sex-matched controls (n = 130) (2.2 ± 3.2 mg/l vs. 1.5 mg/l ± 1.4, respectively, P = 0.037), PTX3 levels were lower in patients with CSCR (10.5 ± 19.9 pg/ml vs. 87.4 ± 73.2 pg/ml, respectively, P < 0.001). There was no significant difference in CRP or PTX3 levels between patients with acute/recurrent and chronic CSCR. Conclusions In patients with CSCR, high CRP and low PTX3 levels suggest a form of low-grade systemic inflammation together with a lack of glucocorticoid pathway activation, raising new hypotheses on the pathophysiology of CSCR. Financial Disclosures The author(s) have no proprietary or commercial interest in any materials discussed in this article.
Collapse
Affiliation(s)
- Elodie Bousquet
- Department of Ophthalmology, Cochin Hospital, Assistance Publique-Hôpitaux de Paris, University of Paris Cité, Paris, France
- Centre de Recherche des Cordeliers, INSERM, University of Paris Cité, Physiopathology of ocular diseases: Therapeutic innovations, Paris, France
| | - Camille Chenevier-Gobeaux
- Service de diagnostic biologique automatisé, Cochin Hospital, Assistance Publique-Hôpitaux de Paris, University of Paris Cité, Paris, France
| | - Thara Jaworski
- Centre de Recherche des Cordeliers, INSERM, University of Paris Cité, Physiopathology of ocular diseases: Therapeutic innovations, Paris, France
| | - Héloïse Torres-Villaros
- Department of Ophthalmology, Cochin Hospital, Assistance Publique-Hôpitaux de Paris, University of Paris Cité, Paris, France
| | - Marta Zola
- Department of Ophthalmology, Cochin Hospital, Assistance Publique-Hôpitaux de Paris, University of Paris Cité, Paris, France
- Centre de Recherche des Cordeliers, INSERM, University of Paris Cité, Physiopathology of ocular diseases: Therapeutic innovations, Paris, France
| | - Irmela Mantel
- Department of Ophthalmology, Jules Gonin Eye Hospital, Fondation Asile des Aveugles, University of Lausanne, Switzerland
| | - Laura Kowalczuk
- Department of Ophthalmology, Jules Gonin Eye Hospital, Fondation Asile des Aveugles, University of Lausanne, Switzerland
| | - Alexandre Matet
- Department of Ophthalmology, Institut Curie, University of Paris Cité, Paris, France
| | - Alejandra Daruich
- Centre de Recherche des Cordeliers, INSERM, University of Paris Cité, Physiopathology of ocular diseases: Therapeutic innovations, Paris, France
- Department of Ophthalmology, Necker Enfants Malades Hospital, Assistance Publique-Hôpitaux de Paris, University of Paris Cité, Paris, France
| | - Min Zhao
- Centre de Recherche des Cordeliers, INSERM, University of Paris Cité, Physiopathology of ocular diseases: Therapeutic innovations, Paris, France
| | - Suzanne Yzer
- Department of Ophthalmology, Rotterdam Eye Hospital, Rotterdam, the Netherlands
| | - Francine Behar-Cohen
- Department of Ophthalmology, Cochin Hospital, Assistance Publique-Hôpitaux de Paris, University of Paris Cité, Paris, France
- Centre de Recherche des Cordeliers, INSERM, University of Paris Cité, Physiopathology of ocular diseases: Therapeutic innovations, Paris, France
- Correspondence: Francine Behar-Cohen, MD, PhD, centre de recherche des cordeliers, 15 rue de l’école de médecine, 75006 Paris, France.
| |
Collapse
|
10
|
Ma Z, Mao C, Jia Y, Yu F, Xu P, Tan Y, Zou QH, Zhou XJ, Kong W, Fu Y. ADAMTS7-Mediated Complement Factor H Degradation Potentiates Complement Activation to Contributing to Renal Injuries. J Am Soc Nephrol 2023; 34:291-308. [PMID: 36735376 PMCID: PMC10103097 DOI: 10.1681/asn.0000000000000004] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 10/31/2022] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND The dysfunction of complement factor H (CFH), the main soluble complement negative regulator, potentiates various complement-induced renal injuries. However, insights into the underlying mechanism of CFH dysfunction remain limited. In this study, we investigated whether extracellular protease-mediated degradation accounts for CFH dysfunction in complement-mediated renal injuries. METHODS An unbiased interactome of lupus mice kidneys identified CFH-binding protease. In vitro cleavage assay clarified CFH degradation. Pristane-induced SLE or renal ischemia-reperfusion (I/R) injury models were used in wild-type and ADAMTS7-/- mice. RESULTS We identified the metalloprotease ADAMTS7 as a CFH-binding protein in lupus kidneys. Moreover, the upregulation of ADAMTS7 correlated with CFH reduction in both lupus mice and patients. Mechanistically, ADAMTS7 is directly bound to CFH complement control protein (CCP) 1-4 domain and degraded CCP 1-7 domain through multiple cleavages. In mice with lupus nephritis or renal I/R injury, ADAMTS7 deficiency alleviated complement activation and related renal pathologies, but without affecting complement-mediated bactericidal activity. Adeno-associated virus-mediated CFH silencing compromised these protective effects of ADAMTS7 knockout against complement-mediated renal injuries in vivo. CONCLUSION ADAMTS7-mediated CFH degradation potentiates complement activation and related renal injuries. ADAMTS7 would be a promising anticomplement therapeutic target that does not increase bacterial infection risk.
Collapse
Affiliation(s)
- Zihan Ma
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Chenfeng Mao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
- Beijing Institute of Biotechnology, Beijing, China
| | - Yiting Jia
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Fang Yu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Ping Xu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Research Unit of Proteomics & Research and Development of New Drugs of Chinese Academy of Medical Sciences, Beijing Institute of Lifeomics, Beijing, China
| | - Ying Tan
- Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Key Laboratory of Renal Disease, Ministry of Health of China, Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
| | - Qing-Hua Zou
- Department of Microbiology & Infectious Disease Center, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Xu-Jie Zhou
- Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Key Laboratory of Renal Disease, Ministry of Health of China, Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
| | - Wei Kong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Yi Fu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| |
Collapse
|
11
|
Laudanski K, Liu D, Gullipalli D, Song WC, Okeke T, Szeto WY. A decline of protective apolipoprotein J and complement factor H concomitant with increase in C5a 3 months after cardiac surgery-Evidence of long-term complement perturbations. Front Cardiovasc Med 2022; 9:983617. [PMID: 36606279 PMCID: PMC9808065 DOI: 10.3389/fcvm.2022.983617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 11/23/2022] [Indexed: 12/24/2022] Open
Abstract
Background Heart surgery results in complement activation with the potential for collateral end-organ damage, especially if the protective elements (complement factor H, Apolipoprotein J) are inadequate. Here, we have investigated if peri-operative stress results in an imbalance between complement activation and its protective mechanisms up to 3 months after heart surgery. Methods 101 patients scheduled for non-emergent cardiac surgery donated blood before the procedure (tbaseline), and 24 h (t24h ), 7 days (t7d ) and 3 months (t3m ) after. Complement activation was measured as a serum level of soluble activated component 5 (sC5a) and soluble terminal complement complex (sTCC). Simultaneously, protective complement factor H (CfH), and apolipoprotein J (ApoJ) were measured. Inflammatory responses were quantified using C-reactive protein (CRP) and interleukin-6 (IL-6). Details regarding anesthesia, intensive care unit (ICU) stay, pre-existing conditions, the incidence of postoperative complications, and mortality were collected from medical records. Results C5a declined at t24h to rebound at t7d and t3m . sTCC was significantly depressed at t24h and returned to baseline at later time points. In contrast, CfH and ApoJ were depressed at t3m . Milieu of complement factors aligned along two longitudinal patterns:cluster#1 (C5a/sTTC continuously increasing and CfH/ApoJ preserved at tbaseline) and cluster#2 (transient sC5a/sTTC increase and progressive decline of CfH). Most patients belonged to cluster #1 at t24h (68%), t7d (74%) and t3m (72%). sTCC correlated with APACHE1h (r 2 =-0.25; p < 0.031) and APACHE24h (r 2 = 0.27; p < 0.049). IL-6 correlated with C5a (r 2 =-0.28; p < 0.042) and sTTC (r 2 =-0.28; p < 0.015). Peri-operative administration of acetaminophen and aspirin altered the complement elements. Prolonged hospital stay correlated with elevated C5a [t (78) = 2.03; p = 0.048] and sTTC serum levels [U (73) = 2.07; p = 0.037]. Patients with stroke had a decreased serum level of C5a at t7d and t3m. Conclusion There is a significant decrease in complement protective factors 3 months after cardiac surgery, while C5a seems to be slightly elevated, suggesting that cardiac surgery affects complement milieu long into recovery.
Collapse
Affiliation(s)
- Krzysztof Laudanski
- Department of Anesthesiology and Critical Care, The University of Pennsylvania, Philadelphia, PA, United States,Department of Neurology, The University of Pennsylvania, Philadelphia, PA, United States,Leonard Davis Institute for Health Economics, The University of Pennsylvania, Philadelphia, PA, United States,*Correspondence: Krzysztof Laudanski,
| | - Da Liu
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Damodar Gullipalli
- Department of Systems Pharmacology and Translational Therapeutics, The University of Pennsylvania, Philadelphia, PA, United States
| | - Wen-Chao Song
- Department of Systems Pharmacology and Translational Therapeutics, The University of Pennsylvania, Philadelphia, PA, United States
| | - Tony Okeke
- Department of Bioengineering, Drexel University, Philadelphia, PA, United States
| | - Wilson Y. Szeto
- Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
12
|
Alic L, Binder CJ, Papac-Milicevic N. The OSE complotype and its clinical potential. Front Immunol 2022; 13:1010893. [PMID: 36248824 PMCID: PMC9561429 DOI: 10.3389/fimmu.2022.1010893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 09/12/2022] [Indexed: 11/17/2022] Open
Abstract
Cellular death, aging, and tissue damage trigger inflammation that leads to enzymatic and non-enzymatic lipid peroxidation of polyunsaturated fatty acids present on cellular membranes and lipoproteins. This results in the generation of highly reactive degradation products, such as malondialdehyde (MDA) and 4-hydroxynonenal (4-HNE), that covalently modify free amino groups of proteins and lipids in their vicinity. These newly generated neoepitopes represent a unique set of damage-associated molecular patterns (DAMPs) associated with oxidative stress termed oxidation-specific epitopes (OSEs). OSEs are enriched on oxidized lipoproteins, microvesicles, and dying cells, and can trigger sterile inflammation. Therefore, prompt recognition and removal of OSEs is required to maintain the homeostatic balance. This is partially achieved by various humoral components of the innate immune system, such as natural IgM antibodies, pentraxins and complement components that not only bind OSEs but in some cases modulate their pro-inflammatory potential. Natural IgM antibodies are potent complement activators, and 30% of them recognize OSEs such as oxidized phosphocholine (OxPC-), 4-HNE-, and MDA-epitopes. Furthermore, OxPC-epitopes can bind the complement-activating pentraxin C-reactive protein, while MDA-epitopes are bound by C1q, C3a, complement factor H (CFH), and complement factor H-related proteins 1, 3, 5 (FHR-1, FHR-3, FHR-5). In addition, CFH and FHR-3 are recruited to 2-(ω-carboxyethyl)pyrrole (CEP), and full-length CFH also possesses the ability to attenuate 4-HNE-induced oxidative stress. Consequently, alterations in the innate humoral defense against OSEs predispose to the development of diseases associated with oxidative stress, as shown for the prototypical OSE, MDA-epitopes. In this mini-review, we focus on the mechanisms of the accumulation of OSEs, the pathophysiological consequences, and the interactions between different OSEs and complement components. Additionally, we will discuss the clinical potential of genetic variants in OSE-recognizing complement proteins – the OSE complotype - in the risk estimation of diseases associated with oxidative stress.
Collapse
Affiliation(s)
- Lejla Alic
- Department of Medical Biochemistry, Faculty of Medicine, University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Christoph J. Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Nikolina Papac-Milicevic
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
- *Correspondence: Nikolina Papac-Milicevic,
| |
Collapse
|
13
|
Laudanski K, Okeke T, Siddiq K, Hajj J, Restrepo M, Gullipalli D, Song WC. A disturbed balance between blood complement protective factors (FH, ApoE) and common pathway effectors (C5a, TCC) in acute COVID-19 and during convalesce. Sci Rep 2022; 12:13658. [PMID: 35953544 PMCID: PMC9366819 DOI: 10.1038/s41598-022-17011-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 07/18/2022] [Indexed: 11/09/2022] Open
Abstract
A complement effect on homeostasis during infection is determined by both cytotoxic (activate complement component 5 (C5a) terminal cytotoxic complex (TCC)), and cytoprotective elements (complement factor H (FH), as well as apolipoprotein E (ApoE)). Here, we investigated the gap in knowledge in their blood milieu during SARS-CoV-2 infection with respect to the viral burden, level of tissue necrosis, and immunological response. 101 patients hospitalized with a PCR-confirmed diagnosis of COVID-19 had blood collected at H1 (48 h), H2 (3-4 Days), H3 (5-7 days), H4 (more than 7 days up to 93 days). Pre-existing conditions, treatment, the incidence of cerebrovascular events (CVA), a history of deep venous thrombosis (DVT) and pulmonary embolism (PE), and mortality was collected using electronic medical records. Plasma C5a, TCC, FH, and ApoE were considered as a complement milieu. Tissue necrosis (HMGB1, RAGE), non-specific inflammatory responses (IL-6, C-reactive protein), overall viral burden (SARS-CoV-2 spike protein), and specific immune responses (IgG, IgA, IgM directed αS- & N-proteins) were assessed simultaneously. C5a remained elevated across all time points, with the peak at 5-7 days. Studied elements of complement coalesced around three clusters: #0 (↑↑↑C5a, ↑↑TCC, ↓↓ApoE), #1 ↑C5a, ↑TCC, ↑↑↑FH); #2 (↑C5a, ↑TCC, ↑FH, ↑↑↑ApoE). The decline in FH and ApoE was a predictor of death, while TCC and C5a correlated with patient length of stay, APACHE, and CRP. Increased levels of C5a (Δ = 122.64; p = 0.0294; data not shown) and diminished levels of FH (Δ = 836,969; p = 0.0285; data not shown) co-existed with CVA incidence. C5a correlated storngly with blood RAGE and HMGB1, but not with viral load and immunological responsiveness. Remdesivir positively affected FH preservation, while convalescent plasma treatment elevated C5a levels. Three clusters of complement activation demonstrated a various milieu of ApoE & FH vs C5a & TCC in COVID-19 patients. Complement activation is linked to increased necrosis markers but not to viral burden or immune system response.
Collapse
Affiliation(s)
- Krzysztof Laudanski
- Department of Anesthesiology and Critical Care, The University of Pennsylvania, JMB 127, 3620 Hamilton Walk, Philadelphia, PA, 19146, USA.
- Leonard Davis Institute for Health Economics, The University of Pennsylvania Colonial Penn Center, 3641 Locust Walk, Philadelphia, PA, 19104, USA.
| | - Tony Okeke
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, USA
| | - Kumal Siddiq
- College of Arts and Sciences, Drexel University, Philadelphia, PA, USA
| | - Jihane Hajj
- School of Nursing, Widener College, Chester, PA, USA
| | - Mariana Restrepo
- College of Arts and Sciences, The University of Pennsylvania, Philadelphia, PA, USA
| | - Damodar Gullipalli
- Department of Systems Pharmacology and Translational Therapeutics, The University of Pennsylvania, Philadelphia, PA, USA
| | - Wen-Chao Song
- Department of Systems Pharmacology and Translational Therapeutics, The University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
14
|
Stravalaci M, Ferrara M, Pathak V, Davi F, Bottazzi B, Mantovani A, Medina RJ, Romano MR, Inforzato A. The Long Pentraxin PTX3 as a New Biomarker and Pharmacological Target in Age-Related Macular Degeneration and Diabetic Retinopathy. Front Pharmacol 2022; 12:811344. [PMID: 35069222 PMCID: PMC8776640 DOI: 10.3389/fphar.2021.811344] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 12/20/2021] [Indexed: 12/12/2022] Open
Abstract
Age related macular degeneration (AMD) and diabetic retinopathy (DR) are multifactorial, neurodegenerative and inflammatory diseases of the eye primarily involving cellular and molecular components of the outer and inner blood-retina barriers (BRB), respectively. Largely contributed by genetic factors, particularly polymorphisms in complement genes, AMD is a paradigm of retinal immune dysregulation. DR, a major complication of diabetes mellitus, typically presents with increased vascular permeability and occlusion of the retinal vasculature that leads, in the proliferative form of the disease, to neovascularization, a pathogenic trait shared with advanced AMD. In spite of distinct etiology and clinical manifestations, both pathologies share common drivers, such as chronic inflammation, either of immune (in AMD) or metabolic (in DR) origin, which initiates and propagates degeneration of the neural retina, yet the underlying mechanisms are still unclear. As a soluble pattern recognition molecule with complement regulatory functions and a marker of vascular damage, long pentraxin 3 (PTX3) is emerging as a novel player in ocular homeostasis and a potential pharmacological target in neurodegenerative disorders of the retina. Physiologically present in the human eye and induced in inflammatory conditions, this protein is strategically positioned at the BRB interface, where it acts as a “molecular trap” for complement, and modulates inflammation both in homeostatic and pathological conditions. Here, we discuss current viewpoints on PTX3 and retinal diseases, with a focus on AMD and DR, the roles therein proposed for this pentraxin, and their implications for the development of new therapeutic strategies.
Collapse
Affiliation(s)
| | | | - Varun Pathak
- School of Medicine, Dentistry, and Biomedical Sciences, Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | | | | | - Alberto Mantovani
- IRCCS Humanitas Research Hospital, Rozzano, Italy.,Department of Biomedical Sciences, Humanitas University, Rozzano, Italy.,The William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Reinhold J Medina
- School of Medicine, Dentistry, and Biomedical Sciences, Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Mario R Romano
- Eye Center, Humanitas Gavazzeni-Castelli, Bergamo, Italy.,Department of Biomedical Sciences, Humanitas University, Rozzano, Italy
| | - Antonio Inforzato
- IRCCS Humanitas Research Hospital, Rozzano, Italy.,Department of Biomedical Sciences, Humanitas University, Rozzano, Italy
| |
Collapse
|
15
|
Schäfer N, Rasras A, Ormenisan DM, Amslinger S, Enzmann V, Jägle H, Pauly D. Complement Factor H-Related 3 Enhanced Inflammation and Complement Activation in Human RPE Cells. Front Immunol 2021; 12:769242. [PMID: 34819935 PMCID: PMC8606654 DOI: 10.3389/fimmu.2021.769242] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 10/18/2021] [Indexed: 12/14/2022] Open
Abstract
Complement Factor H-Related 3 (FHR-3) is a major regulator of the complement system, which is associated with different diseases, such as age-related macular degeneration (AMD). However, the non-canonical local, cellular functions of FHR-3 remained poorly understood. Here, we report that FHR-3 bound to oxidative stress epitopes and competed with FH for interaction. Furthermore, FHR-3 was internalized by viable RPE cells and modulated time-dependently complement component (C3, FB) and receptor (C3aR, CR3) expression of human RPE cells. Independently of any external blood-derived proteins, complement activation products were detected. Anaphylatoxin C3a was visualized in treated cells and showed a translocation from the cytoplasm to the cell membrane after FHR-3 exposure. Subsequently, FHR-3 induced a RPE cell dependent pro-inflammatory microenvironment. Inflammasome NLRP3 activation and pro-inflammatory cytokine secretion of IL-1ß, IL-18, IL-6 and TNF-α were induced after FHR-3-RPE interaction. Our previously published monoclonal anti-FHR-3 antibody, which was chimerized to reduce immunogenicity, RETC-2-ximab, ameliorated the effect of FHR-3 on ARPE-19 cells. Our studies suggest FHR-3 as an exogenous trigger molecule for the RPE cell "complosome" and as a putative target for a therapeutic approach for associated degenerative diseases.
Collapse
Affiliation(s)
- Nicole Schäfer
- Department of Ophthalmology, University Hospital Regensburg, Regensburg, Germany
- Department of Orthopaedic Surgery, Experimental Orthopaedics, Centre for Medical Biotechnology (ZMB), University of Regensburg, Regensburg, Germany
| | - Anas Rasras
- Chemistry Department, Al-Balqa Applied University, Al-Salt, Jordan
- Institute of Organic Chemistry, University of Regensburg, Regensburg, Germany
| | - Delia M. Ormenisan
- Department of Ophthalmology, University Hospital Regensburg, Regensburg, Germany
| | - Sabine Amslinger
- Institute of Organic Chemistry, University of Regensburg, Regensburg, Germany
| | - Volker Enzmann
- Department of Ophthalmology, University Hospital of Bern and Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Herbert Jägle
- Department of Ophthalmology, University Hospital Regensburg, Regensburg, Germany
| | - Diana Pauly
- Department of Ophthalmology, University Hospital Regensburg, Regensburg, Germany
- Experimental Ophthalmology, Philipps-University Marburg, Marburg, Germany
| |
Collapse
|
16
|
Pathogenic Effects of Mineralocorticoid Pathway Activation in Retinal Pigment Epithelium. Int J Mol Sci 2021; 22:ijms22179618. [PMID: 34502527 PMCID: PMC8431771 DOI: 10.3390/ijms22179618] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 08/25/2021] [Accepted: 08/30/2021] [Indexed: 12/17/2022] Open
Abstract
Glucocorticoids are amongst the most used drugs to treat retinal diseases of various origins. Yet, the transcriptional regulations induced by glucocorticoid receptor (GR) and mineralocorticoid receptor (MR) activation in retinal pigment epithelium cells (RPE) that form the outer blood-retina barrier are unknown. Levels of endogenous corticoids, ligands for MR and GR, were measured in human ocular media. Human RPE cells derived from induced pluripotent stem cells (iRPE) were used to analyze the pan-transcriptional regulations induced by aldosterone-an MR-specific agonist, or cortisol or cortisol + RU486-a GR antagonist. The retinal phenotype of transgenic mice that overexpress the human MR (P1.hMR) was analyzed. In the human eye, the main ligand for GR and MR is cortisol. The iRPE cells express functional GR and MR. The subset of genes regulated by aldosterone and by cortisol + RU-486, and not by cortisol alone, mimics an imbalance toward MR activation. They are involved in extracellular matrix remodeling (CNN1, MGP, AMTN), epithelial-mesenchymal transition, RPE cell proliferation and migration (ITGB3, PLAUR and FOSL1) and immune balance (TNFSF18 and PTX3). The P1.hMR mice showed choroidal vasodilation, focal alteration of the RPE/choroid interface and migration of RPE cells together with RPE barrier function alteration, similar to human retinal diseases within the pachychoroid spectrum. RPE is a corticosteroid-sensitive epithelium. MR pathway activation in the RPE regulates genes involved in barrier function, extracellular matrix, neural regulation and epithelial differentiation, which could contribute to retinal pathology.
Collapse
|
17
|
Kim BJ, Mastellos DC, Li Y, Dunaief JL, Lambris JD. Targeting complement components C3 and C5 for the retina: Key concepts and lingering questions. Prog Retin Eye Res 2021; 83:100936. [PMID: 33321207 PMCID: PMC8197769 DOI: 10.1016/j.preteyeres.2020.100936] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 12/07/2020] [Accepted: 12/09/2020] [Indexed: 12/13/2022]
Abstract
Age-related macular degeneration (AMD) remains a major cause of legal blindness, and treatment for the geographic atrophy form of AMD is a significant unmet need. Dysregulation of the complement cascade is thought to be instrumental for AMD pathophysiology. In particular, C3 and C5 are pivotal components of the complement cascade and have become leading therapeutic targets for AMD. In this article, we discuss C3 and C5 in detail, including their roles in AMD, biochemical and structural aspects, locations of expression, and the functions of C3 and C5 fragments. Further, the article critically reviews developing therapeutics aimed at C3 and C5, underscoring the potential effects of broad inhibition of complement at the level of C3 versus more specific inhibition at C5. The relationships of complement biology to the inflammasome and microglia/macrophage activity are highlighted. Concepts of C3 and C5 biology will be emphasized, while we point out questions that need to be settled and directions for future investigations.
Collapse
Affiliation(s)
- Benjamin J Kim
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | | | - Yafeng Li
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Joshua L Dunaief
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - John D Lambris
- Department of Laboratory Medicine and Pathology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
18
|
Immunological Aspects of Age-Related Macular Degeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1256:143-189. [PMID: 33848001 DOI: 10.1007/978-3-030-66014-7_6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Increasing evidence over the past two decades points to a pivotal role for immune mechanisms in age-related macular degeneration (AMD) pathobiology. In this chapter, we will explore immunological aspects of AMD, with a specific focus on how immune mechanisms modulate clinical phenotypes of disease and severity and how components of the immune system may serve as triggers for disease progression in both dry and neovascular AMD. We will briefly review the biology of the immune system, defining the role of immune mechanisms in chronic degenerative disease and differentiating from immune responses to acute injury or infection. We will explore current understanding of the roles of innate immunity (especially macrophages), antigen-specific immunity (T cells, B cells, and autoimmunity), immune amplifications systems, especially complement activity and the NLRP3 inflammasome, in the pathogenesis of both dry and neovascular AMD, reviewing data from pathology, experimental animal models, and clinical studies of AMD patients. We will also assess how interactions between the immune system and infectious pathogens could potentially modulate AMD pathobiology via alterations in in immune effector mechanisms. We will conclude by reviewing the paradigm of "response to injury," which provides a means to integrate various immunologic mechanisms along with nonimmune mechanisms of tissue injury and repair as a model to understand the pathobiology of AMD.
Collapse
|
19
|
Stravalaci M, Davi F, Parente R, Gobbi M, Bottazzi B, Mantovani A, Day AJ, Clark SJ, Romano MR, Inforzato A. Control of Complement Activation by the Long Pentraxin PTX3: Implications in Age-Related Macular Degeneration. Front Pharmacol 2020; 11:591908. [PMID: 33324220 PMCID: PMC7725797 DOI: 10.3389/fphar.2020.591908] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 10/28/2020] [Indexed: 12/28/2022] Open
Abstract
Dysregulation of the complement system is central to age-related macular degeneration (AMD), the leading cause of blindness in the developed world. Most of the genetic variation associated with AMD resides in complement genes, with the greatest risk associated with polymorphisms in the complement factor H (CFH) gene; factor H (FH) is the major inhibitor of the alternative pathway (AP) of complement that specifically targets C3b and the AP C3 convertase. Long pentraxin 3 (PTX3) is a soluble pattern recognition molecule that has been proposed to inhibit AP activation via recruitment of FH. Although present in the human retina, if and how PTX3 plays a role in AMD is still unclear. In this work we demonstrated the presence of PTX3 in the human vitreous and studied the PTX3-FH-C3b crosstalk and its effects on complement activation in a model of retinal pigment epithelium (RPE). RPE cells cultured in inflammatory AMD-like conditions overexpressed the PTX3 protein, and up-regulated AP activating genes. PTX3 bound RPE cells in a physiological setting, however this interaction was reduced in inflammatory conditions, whereby PTX3 had no complement-inhibiting activity on inflamed RPE. However, on non-cellular surfaces, PTX3 formed a stable ternary complex with FH and C3b that acted as a “hot spot” for complement inhibition. Our findings suggest a protective role for PTX3 in response to complement dysregulation in AMD and point to a novel mechanism of complement regulation by this pentraxin with potential implications in pathology and pharmacology of AMD.
Collapse
Affiliation(s)
- Matteo Stravalaci
- Department of Biomedical Sciences, Humanitas University, Milan, Italy.,Humanitas Clinical and Research Center IRCCS, Milan, Italy
| | - Francesca Davi
- Humanitas Clinical and Research Center IRCCS, Milan, Italy
| | | | - Marco Gobbi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | | | - Alberto Mantovani
- Department of Biomedical Sciences, Humanitas University, Milan, Italy.,Humanitas Clinical and Research Center IRCCS, Milan, Italy.,The William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Anthony J Day
- Wellcome Trust Centre for Cell-Matrix Research and Lydia Becker Institute of Immunology and Inflammation, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Simon J Clark
- Universitäts-Augenklinik Tübingen, Eberhard Karls University of Tübingen, Tübingen, Germany.,The Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Mario R Romano
- Department of Biomedical Sciences, Humanitas University, Milan, Italy.,Eye Center, Humanitas Gavazzeni-Castelli, Bergamo, Italy
| | - Antonio Inforzato
- Department of Biomedical Sciences, Humanitas University, Milan, Italy.,Humanitas Clinical and Research Center IRCCS, Milan, Italy
| |
Collapse
|
20
|
Genc AM, Makia MS, Sinha T, Conley SM, Al-Ubaidi MR, Naash MI. Retbindin: A riboflavin Binding Protein, Is Critical for Photoreceptor Homeostasis and Survival in Models of Retinal Degeneration. Int J Mol Sci 2020; 21:ijms21218083. [PMID: 33138244 PMCID: PMC7662319 DOI: 10.3390/ijms21218083] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/19/2020] [Accepted: 10/23/2020] [Indexed: 12/12/2022] Open
Abstract
The large number of inherited retinal disease genes (IRD), including the photopigment rhodopsin and the photoreceptor outer segment (OS) structural component peripherin 2 (PRPH2), has prompted interest in identifying common cellular mechanisms involved in degeneration. Although metabolic dysregulation has been shown to play an important role in the progression of the disease etiology, identifying a common regulator that can preserve the metabolic ecosystem is needed for future development of neuroprotective treatments. Here, we investigated whether retbindin (RTBDN), a rod-specific protein with riboflavin binding capability, and a regulator of riboflavin-derived cofactors flavin mononucleotide (FMN) and flavin adenine dinucleotide (FAD), is protective to the retina in different IRD models; one carrying the P23H mutation in rhodopsin (which causes retinitis pigmentosa) and one carrying the Y141C mutation in Prph2 (which causes a blended cone-rod dystrophy). RTBDN levels are significantly upregulated in both the rhodopsin (Rho)P23H/+ and Prph2Y141C/+ retinas. Rod and cone structural and functional degeneration worsened in models lacking RTBDN. In addition, removing Rtbdn worsened other phenotypes, such as fundus flecking. Retinal flavin levels were reduced in RhoP23H/+/Rtbdn−/− and Prph2Y141C/+/Rtbdn−/− retinas. Overall, these findings suggest that RTBDN may play a protective role during retinal degenerations that occur at varying rates and due to varying disease mechanisms.
Collapse
Affiliation(s)
- Ayse M. Genc
- Department of Biomedical Engineering, University of Houston, Houston, TX 77204, USA; (A.M.G.); (M.S.M.); (T.S.)
| | - Mustafa S. Makia
- Department of Biomedical Engineering, University of Houston, Houston, TX 77204, USA; (A.M.G.); (M.S.M.); (T.S.)
| | - Tirthankar Sinha
- Department of Biomedical Engineering, University of Houston, Houston, TX 77204, USA; (A.M.G.); (M.S.M.); (T.S.)
| | - Shannon M. Conley
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
- Oklahoma Center for Neurosciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Muayyad R. Al-Ubaidi
- Department of Biomedical Engineering, University of Houston, Houston, TX 77204, USA; (A.M.G.); (M.S.M.); (T.S.)
- College of Optometry, University of Houston, Houston, TX 77204, USA
- Department of Biology and Biochemistry, University of Houston, TX 77204, USA
- Correspondence: (M.R.A.-U.); (M.I.N.); Tel.: +1-713-743-1651 (M.R.A.-U. & M.I.N.); Fax: +1-713-743-0226 (M.R.A.-U. & M.I.N.)
| | - Muna I. Naash
- Department of Biomedical Engineering, University of Houston, Houston, TX 77204, USA; (A.M.G.); (M.S.M.); (T.S.)
- College of Optometry, University of Houston, Houston, TX 77204, USA
- Department of Biology and Biochemistry, University of Houston, TX 77204, USA
- Correspondence: (M.R.A.-U.); (M.I.N.); Tel.: +1-713-743-1651 (M.R.A.-U. & M.I.N.); Fax: +1-713-743-0226 (M.R.A.-U. & M.I.N.)
| |
Collapse
|
21
|
Cheng X, He D, Liao C, Lin S, Tang L, Wang YL, Hu J, Li W, Liu Z, Wu Y, Liao Y. IL-1/IL-1R signaling induced by all-trans-retinal contributes to complement alternative pathway activation in retinal pigment epithelium. J Cell Physiol 2020; 236:3660-3674. [PMID: 33034385 DOI: 10.1002/jcp.30103] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 09/27/2020] [Accepted: 09/29/2020] [Indexed: 12/23/2022]
Abstract
The underlying mechanisms of complement activation in Stargardt disease type 1 (STGD1) and age-related macular degeneration (AMD) are not fully understood. Overaccumulation of all-trans-retinal (atRAL) has been proposed as the pathogenic factor in both diseases. By incubating retinal pigment epithelium (RPE) cells with atRAL, we showed that C5b-9 membrane attack complexes (MACs) were generated mainly through complement alternative pathway. An increase in complement factor B (CFB) expression as well as downregulation of complement regulatory proteins CD46, CD55, CD59, and CFH were observed in RPE cells after atRAL treatment. Furthermore, interleukin-1β production was provoked in both atRAL-treated RPE cells and microglia/macrophages. Coincubation of RPE cells with interleukin-1 receptor antagonist (IL1Ra) and atRAL ameliorated complement activation and downregulated CFB expression by attenuating both p38 and c-Jun N-terminal kinase (JNK) signaling pathways. Our findings demonstrate that atRAL induces an autocrine/paracrine IL-1/IL-1R signaling to promote complement alternative pathway activation in RPE cells and provide a novel perspective on the pathomechanism of macular degeneration.
Collapse
Affiliation(s)
- Xinxuan Cheng
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Eye Institute of Xiamen University, Xiamen University, Xiamen, China.,Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, China.,Department of Ophthalmology, Xiamen University Affiliated Xiamen Eye Center, Xiamen, China
| | - Danxue He
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Eye Institute of Xiamen University, Xiamen University, Xiamen, China.,Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, China.,Department of Ophthalmology, Xiamen University Affiliated Xiamen Eye Center, Xiamen, China
| | - Chunyan Liao
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Eye Institute of Xiamen University, Xiamen University, Xiamen, China.,Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, China.,Department of Ophthalmology, Xiamen University Affiliated Xiamen Eye Center, Xiamen, China
| | - Sijie Lin
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Eye Institute of Xiamen University, Xiamen University, Xiamen, China.,Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, China.,Department of Ophthalmology, Xiamen University Affiliated Xiamen Eye Center, Xiamen, China
| | - Liying Tang
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Eye Institute of Xiamen University, Xiamen University, Xiamen, China.,Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, China.,Department of Ophthalmology, Xiamen University Affiliated Xiamen Eye Center, Xiamen, China
| | - Yuan-Liang Wang
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, China.,Section of Molecular Biology, University of California, San Diego, La Jolla, California, USA
| | - Jiaoyue Hu
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Eye Institute of Xiamen University, Xiamen University, Xiamen, China.,Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, China.,Department of Ophthalmology, Xiamen University Affiliated Xiamen Eye Center, Xiamen, China
| | - Wei Li
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Eye Institute of Xiamen University, Xiamen University, Xiamen, China.,Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, China.,Department of Ophthalmology, Xiamen University Affiliated Xiamen Eye Center, Xiamen, China
| | - Zuguo Liu
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Eye Institute of Xiamen University, Xiamen University, Xiamen, China.,Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, China.,Department of Ophthalmology, Xiamen University Affiliated Xiamen Eye Center, Xiamen, China
| | - Yalin Wu
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Eye Institute of Xiamen University, Xiamen University, Xiamen, China.,Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, China.,Department of Ophthalmology, Xiamen University Affiliated Xiamen Eye Center, Xiamen, China
| | - Yi Liao
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Eye Institute of Xiamen University, Xiamen University, Xiamen, China.,Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, China.,Department of Ophthalmology, Xiamen University Affiliated Xiamen Eye Center, Xiamen, China
| |
Collapse
|
22
|
Schäfer N, Wolf HN, Enzbrenner A, Schikora J, Reichenthaler M, Enzmann V, Pauly D. Properdin Modulates Complement Component Production in Stressed Human Primary Retinal Pigment Epithelium Cells. Antioxidants (Basel) 2020; 9:E793. [PMID: 32859013 PMCID: PMC7555107 DOI: 10.3390/antiox9090793] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/15/2020] [Accepted: 08/22/2020] [Indexed: 12/16/2022] Open
Abstract
The retinal pigment epithelium (RPE) maintains visual function and preserves structural integrity of the retina. Chronic dysfunction of the RPE is associated with retinal degeneration, including age-related macular degeneration (AMD). The AMD pathogenesis includes both increased oxidative stress and complement dysregulation. Physiological sources of oxidative stress in the retina are well known, while complement sources and regulation are still under debate. Using human primary RPE (hpRPE) cells, we have established a model to investigate complement component expression on transcript and protein level in AMD-risk and non-risk hpRPE cells. We evaluated the effect of properdin, a complement stabilizer, on the hpRPE cell-dependent complement profile exposed to oxidative stress. hpRPE cells expressed complement components, receptors and regulators. Complement proteins were also stored and secreted by hpRPE cells. We associated AMD-risk single nucleotide polymorphisms with an increased secretion of complement factors D (CFD) and I (CFI). Furthermore, we detected hpRPE cell-associated complement activation products (C3a, C5a) independent of any extracellularly added complement system. Exogenous properdin increased the mRNA expression of CFI and CFD, but decreased levels of complement components (C1Q, C3), receptors (C3AR, C5AR1, CD11B) and inflammation-associated transcripts (NLRP3, IL1B) in hpRPE cells exposed to oxidative stress. This properdin effect was time-dependently counter regulated. In conclusion, our data unveiled a local, genotype-associated complement component production in hpRPE cells, regulated by exogenous properdin. The local complement production and activation via blood-independent mechanisms can be a new therapeutic target for AMD.
Collapse
Affiliation(s)
- Nicole Schäfer
- Experimental Ophthalmology, Eye clinic, University Hospital Regensburg, 93053 Regensburg, Germany; (N.S.); (H.N.W.); (A.E.); (J.S.); (M.R.)
| | - Hannah N. Wolf
- Experimental Ophthalmology, Eye clinic, University Hospital Regensburg, 93053 Regensburg, Germany; (N.S.); (H.N.W.); (A.E.); (J.S.); (M.R.)
| | - Anne Enzbrenner
- Experimental Ophthalmology, Eye clinic, University Hospital Regensburg, 93053 Regensburg, Germany; (N.S.); (H.N.W.); (A.E.); (J.S.); (M.R.)
| | - Juliane Schikora
- Experimental Ophthalmology, Eye clinic, University Hospital Regensburg, 93053 Regensburg, Germany; (N.S.); (H.N.W.); (A.E.); (J.S.); (M.R.)
| | - Maria Reichenthaler
- Experimental Ophthalmology, Eye clinic, University Hospital Regensburg, 93053 Regensburg, Germany; (N.S.); (H.N.W.); (A.E.); (J.S.); (M.R.)
| | - Volker Enzmann
- Department of Ophthalmology, University Hospital of Bern, University of Bern, 3010 Bern, Switzerland;
- Department of Biomedical Research, University of Bern, 3010 Bern, Switzerland
| | - Diana Pauly
- Experimental Ophthalmology, Eye clinic, University Hospital Regensburg, 93053 Regensburg, Germany; (N.S.); (H.N.W.); (A.E.); (J.S.); (M.R.)
| |
Collapse
|
23
|
Kang Y, Yu Y, Lu L. The Role of Pentraxin 3 in Aspergillosis: Reality and Prospects. MYCOBIOLOGY 2020; 48:1-8. [PMID: 32158600 PMCID: PMC7048186 DOI: 10.1080/12298093.2020.1722576] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Revised: 01/10/2020] [Accepted: 01/23/2020] [Indexed: 02/08/2023]
Abstract
Pentraxin 3 (PTX3) is a soluble pattern recognition receptor (PRR), which is produced by several kinds of cells, such as neutrophils, dendritic cells, macrophages, and epithelial cells. PTX3 is known to play an important protective effect against Aspergillus. Genetic linkage in gene-targeted mice and human PTX3 plays a non-redundant role in the immune protection against specific pathogens, especially Aspergillus. Recent studies have shown that the polymorphism of PTX3 is associated with increased susceptibility to invasive aspergillosis (IA). In this review, we provide an overview of these studies that underline the potential of PTX3 in diagnosis and therapy of IA.
Collapse
Affiliation(s)
- Yuening Kang
- Department of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yuetian Yu
- Department of Critical Care Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Liangjing Lu
- Department of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
24
|
Aerobic Training Down-Regulates Pentraxin 3 and Pentraxin 3/Toll-Like Receptor 4 Ratio, Irrespective of Oxidative Stress Response, in Elderly Subjects. Antioxidants (Basel) 2020; 9:antiox9020110. [PMID: 32012711 PMCID: PMC7070734 DOI: 10.3390/antiox9020110] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Revised: 01/21/2020] [Accepted: 01/23/2020] [Indexed: 01/14/2023] Open
Abstract
Reactive oxygen and nitrogen species-mediated cellular aging has been linked to diseases such as atherothrombosis and cancer. Although pentraxin 3 (PTX3) is associated with aging-related diseases via TLR4-dependent anti-inflammatory effects, its relationship with oxidative stress in aging remains to be elucidated. Exercise is proposed as the key intervention for health maintenance in the elderly. This study aimed to examine the association of PTX3 levels with changes in oxidative stress in both plasma and peripheral blood mononuclear cells (PBMCs), following aerobic training in elderly adults. Nine trained and five controls participated in an eight-week aerobic training protocol. Enzyme-linked immunosorbent assay (ELISA) and Western blot analyses were used to determine PTX3, toll-like receptor 4 (TLR4), and levels of oxidative stress biomarkers [3-nitrotyrosine (3NT), 4-hydroxynonenal (4-HNE), glutathione (GSH), protein carbonyl (PC), reactive oxygen/ nitrogen species (ROS/RNS), and trolox equivalent antioxidant capacity (TEAC)] in plasma and/or PBMCs. Results showed a down-regulation of PTX3 expression in PBMCs following aerobic training, along with decreased PTX3/TLR4 ratios. Oxidative stress responses in PBMCs remained unchanged with the exercise protocol. Comparable levels of plasma PTX3 and oxidative stress biomarkers were observed in trained vs. control groups. No correlation was found between PTX3 and any oxidative stress biomarkers following training. These findings demonstrated the down-regulation of PTX3 and PTX3/TLR4 ratio, irrespective of oxidative stress response, in elderly adults following eight weeks of aerobic training.
Collapse
|
25
|
Lipecz A, Miller L, Kovacs I, Czakó C, Csipo T, Baffi J, Csiszar A, Tarantini S, Ungvari Z, Yabluchanskiy A, Conley S. Microvascular contributions to age-related macular degeneration (AMD): from mechanisms of choriocapillaris aging to novel interventions. GeroScience 2019; 41:813-845. [PMID: 31797238 PMCID: PMC6925092 DOI: 10.1007/s11357-019-00138-3] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 11/12/2019] [Indexed: 12/13/2022] Open
Abstract
Aging of the microcirculatory network plays a central role in the pathogenesis of a wide range of age-related diseases, from heart failure to Alzheimer's disease. In the eye, changes in the choroid and choroidal microcirculation (choriocapillaris) also occur with age, and these changes can play a critical role in the pathogenesis of age-related macular degeneration (AMD). In order to develop novel treatments for amelioration of choriocapillaris aging and prevention of AMD, it is essential to understand the cellular and functional changes that occur in the choroid and choriocapillaris during aging. In this review, recent advances in in vivo analysis of choroidal structure and function in AMD patients and patients at risk for AMD are discussed. The pathophysiological roles of fundamental cellular and molecular mechanisms of aging including oxidative stress, mitochondrial dysfunction, and impaired resistance to molecular stressors in the choriocapillaris are also considered in terms of their contribution to the pathogenesis of AMD. The pathogenic roles of cardiovascular risk factors that exacerbate microvascular aging processes, such as smoking, hypertension, and obesity as they relate to AMD and choroid and choriocapillaris changes in patients with these cardiovascular risk factors, are also discussed. Finally, future directions and opportunities to develop novel interventions to prevent/delay AMD by targeting fundamental cellular and molecular aging processes are presented.
Collapse
Affiliation(s)
- Agnes Lipecz
- Translational Geroscience Laboratory, Center for Geroscience and Healthy Brain Aging/Reynolds Oklahoma Center on Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Vascular Cognitive Impairment and Neurodegeneration Program, Center for Geroscience and Healthy Brain Aging/Reynolds Oklahoma Center on Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Ophthalmology, Josa Andras Hospital, Nyiregyhaza, Hungary
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- Department of Ophthalmology, Semmelweis University, Budapest, Hungary
| | - Lauren Miller
- Vascular Cognitive Impairment and Neurodegeneration Program, Center for Geroscience and Healthy Brain Aging/Reynolds Oklahoma Center on Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Cell Biology, University of Oklahoma Health Sciences Center, 940 Stanton L. Young Blvd. BMSB553, Oklahoma City, OK, 73104, USA
| | - Illes Kovacs
- Vascular Cognitive Impairment and Neurodegeneration Program, Center for Geroscience and Healthy Brain Aging/Reynolds Oklahoma Center on Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Ophthalmology, Semmelweis University, Budapest, Hungary
- Department of Ophthalmology, Weill Cornell Medical College, New York City, NY, USA
| | - Cecília Czakó
- Department of Ophthalmology, Semmelweis University, Budapest, Hungary
| | - Tamas Csipo
- Translational Geroscience Laboratory, Center for Geroscience and Healthy Brain Aging/Reynolds Oklahoma Center on Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Vascular Cognitive Impairment and Neurodegeneration Program, Center for Geroscience and Healthy Brain Aging/Reynolds Oklahoma Center on Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- International Training Program in Geroscience, Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Judit Baffi
- Vascular Cognitive Impairment and Neurodegeneration Program, Center for Geroscience and Healthy Brain Aging/Reynolds Oklahoma Center on Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Anna Csiszar
- Translational Geroscience Laboratory, Center for Geroscience and Healthy Brain Aging/Reynolds Oklahoma Center on Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Vascular Cognitive Impairment and Neurodegeneration Program, Center for Geroscience and Healthy Brain Aging/Reynolds Oklahoma Center on Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- International Training Program in Geroscience, Theoretical Medicine Doctoral School, University of Szeged, Szeged, Hungary
| | - Stefano Tarantini
- Translational Geroscience Laboratory, Center for Geroscience and Healthy Brain Aging/Reynolds Oklahoma Center on Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Vascular Cognitive Impairment and Neurodegeneration Program, Center for Geroscience and Healthy Brain Aging/Reynolds Oklahoma Center on Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- International Training Program in Geroscience, Theoretical Medicine Doctoral School, University of Szeged, Szeged, Hungary
| | - Zoltan Ungvari
- Translational Geroscience Laboratory, Center for Geroscience and Healthy Brain Aging/Reynolds Oklahoma Center on Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Vascular Cognitive Impairment and Neurodegeneration Program, Center for Geroscience and Healthy Brain Aging/Reynolds Oklahoma Center on Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- International Training Program in Geroscience, Theoretical Medicine Doctoral School, University of Szeged, Szeged, Hungary
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Andriy Yabluchanskiy
- Translational Geroscience Laboratory, Center for Geroscience and Healthy Brain Aging/Reynolds Oklahoma Center on Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Vascular Cognitive Impairment and Neurodegeneration Program, Center for Geroscience and Healthy Brain Aging/Reynolds Oklahoma Center on Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Shannon Conley
- Vascular Cognitive Impairment and Neurodegeneration Program, Center for Geroscience and Healthy Brain Aging/Reynolds Oklahoma Center on Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Department of Cell Biology, University of Oklahoma Health Sciences Center, 940 Stanton L. Young Blvd. BMSB553, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
26
|
Hwang N, Kwon MY, Woo JM, Chung SW. Oxidative Stress-Induced Pentraxin 3 Expression Human Retinal Pigment Epithelial Cells is Involved in the Pathogenesis of Age-Related Macular Degeneration. Int J Mol Sci 2019; 20:ijms20236028. [PMID: 31795454 PMCID: PMC6928709 DOI: 10.3390/ijms20236028] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 11/23/2019] [Accepted: 11/27/2019] [Indexed: 01/07/2023] Open
Abstract
(1) Background: Age-related macular degeneration (AMD) is closely related with retinal pigment epithelial (RPE) cell dysfunction. Although the exact pathogenesis of AMD remains largely unknown, oxidative stress-induced RPE damage is believed to be one of the primary causes. We investigated the molecular mechanisms of pentraxin 3 (PTX3) expression and its biological functions during oxidative injury. (2) Methods: Using enzyme-linked immunosorbent assays and real-time reverse transcription-polymerase chain reaction, we analyzed mRNA and protein levels of PTX3 in the presence or absence of oxidative stress inducer, sodium iodate (NaIO3), in primary human H-RPE and ARPE-19 cells. Furthermore, we assessed cell death, antioxidant enzyme expression, and AMD-associated gene expression to determine the biological functions of PTX3 under oxidative stress. (3) Results: NaIO3 increased PTX3 expression, in a dose- and time-dependent manner, in H-RPE and ARPE-19 cells. We found phosphorylated Akt, a downstream target of the PI3 kinase pathway, phosphor- mitogen-activated protein kinase kinase 1/2 (ERK), and intracellular reactive oxygen species (ROS) were predominantly induced by NaIO3. NaIO3-induced PTX3 expression was decreased in the presence of phosphoinositide 3 (PI3) kinase inhibitors, ERK inhibitors, and ROS scavengers. Furthermore, NaIO3 enhanced mRNA expression of antioxidant enzymes such as glucose-6-phosphate dehydrogenase (G6PDH), catalase (CAT), and glutathione S-reductase (GSR) in the control shRNA expressing RPE cells, but not in hPTX3 shRNA expressing RPE cells. Interestingly, NaIO3 did not induce mRNA expression of AMD marker genes, such as complement factor I (CFI), complement factor H (CFH), apolipoprotein E (APOE), and toll-like receptor 4 (TLR4) in hPTX3 shRNA expressing RPE cells. 4) Conclusions: These results suggest that PTX3 accelerates RPE cell death and might be involved in AMD development in the presence of oxidative stress.
Collapse
Affiliation(s)
- Narae Hwang
- School of Biological Sciences, College of Natural Sciences, University of Ulsan, 93 Daehak-ro, Ulsan 44610, Korea; (N.H.); (M.-Y.K.)
| | - Min-Young Kwon
- School of Biological Sciences, College of Natural Sciences, University of Ulsan, 93 Daehak-ro, Ulsan 44610, Korea; (N.H.); (M.-Y.K.)
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Je Moon Woo
- Department of Ophthalmology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan 44033, Korea;
| | - Su Wol Chung
- School of Biological Sciences, College of Natural Sciences, University of Ulsan, 93 Daehak-ro, Ulsan 44610, Korea; (N.H.); (M.-Y.K.)
- Correspondence: ; Tel.: +82-52-259-2353; Fax: +82-52-259-1694
| |
Collapse
|
27
|
Wu XY, Li KT, Yang HX, Yang B, Lu X, Zhao LD, Fei YY, Chen H, Wang L, Li J, Peng LY, Zheng WJ, Hou Y, Jiang Y, Shi Q, Zhang W, Zhang FC, Zhang JM, Huang B, He W, Zhang X. Complement C1q synergizes with PTX3 in promoting NLRP3 inflammasome over-activation and pyroptosis in rheumatoid arthritis. J Autoimmun 2019; 106:102336. [PMID: 31601476 DOI: 10.1016/j.jaut.2019.102336] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 09/23/2019] [Accepted: 09/23/2019] [Indexed: 11/27/2022]
Abstract
Excessive inflammatory cytokines play crucial roles in the pathogenesis of rheumatoid arthritis (RA), however, the underlying mechanism remains unclear. In this study, we demonstrated that pentaxin 3 (PTX3), an essential component of innate immunity, was elevated in RA and preferentially bound to CD14+ monocytes. C1q promoted the binding and resulted in increased cell proliferation, activation and caspase-1-related late apoptotic cells (7-AAD+annexin V+), as well as enhanced release of inflammatory cytokines including TNF-α, IL-1β and IL-6. Serum from RA patients, compared with healthy controls, induced gasdermin D (GSDMD)-dependent pyroptosis in monocytes, and this ability was associated with disease activity. Moreover, PTX3 synergized with C1q to promote pyroptosis in RA-serum pre-incubated monocytes by coordinately enhancing NLRP3 inflammasome over-activation and inducing GSDMD cleavage, cell swelling with large bubbles, caspase-1-dependent cell death and inflammatory cytokine release including IL-6. On the other hand, IL-6 promoted PTX3 plus C1q-induced pyroptosis in both normal and RA serum pre-incubated monocytes. These findings collectively implicated an important role of IL-6 in driving PTX3 plus C1q-mediated pyroptosis in RA and shed lights on a potential new treatment strategy targeting pyroptosis-mediated persistent inflammatory cytokine release.
Collapse
Affiliation(s)
- Xun-Yao Wu
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, The Ministry of Education Key Laboratory, Beijing, 100730, China
| | - Ke-Tian Li
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, The Ministry of Education Key Laboratory, Beijing, 100730, China
| | - Hua-Xia Yang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, The Ministry of Education Key Laboratory, Beijing, 100730, China
| | - Bo Yang
- Department of Orthopaedics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Xin Lu
- Department of Orthopaedics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Li-Dan Zhao
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, The Ministry of Education Key Laboratory, Beijing, 100730, China
| | - Yun-Yun Fei
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, The Ministry of Education Key Laboratory, Beijing, 100730, China
| | - Hua Chen
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, The Ministry of Education Key Laboratory, Beijing, 100730, China
| | - Li Wang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, The Ministry of Education Key Laboratory, Beijing, 100730, China
| | - Jing Li
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, The Ministry of Education Key Laboratory, Beijing, 100730, China
| | - Ling-Yi Peng
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, The Ministry of Education Key Laboratory, Beijing, 100730, China
| | - Wen-Jie Zheng
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, The Ministry of Education Key Laboratory, Beijing, 100730, China
| | - Yong Hou
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, The Ministry of Education Key Laboratory, Beijing, 100730, China
| | - Ying Jiang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, The Ministry of Education Key Laboratory, Beijing, 100730, China
| | - Qun Shi
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, The Ministry of Education Key Laboratory, Beijing, 100730, China
| | - Wen Zhang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, The Ministry of Education Key Laboratory, Beijing, 100730, China
| | - Feng-Chun Zhang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, The Ministry of Education Key Laboratory, Beijing, 100730, China
| | - Jian-Min Zhang
- Department of Immunology & National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College, Beijing, 100005, China
| | - Bo Huang
- Department of Immunology & National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College, Beijing, 100005, China
| | - Wei He
- Department of Immunology & National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College, Beijing, 100005, China
| | - Xuan Zhang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, The Ministry of Education Key Laboratory, Beijing, 100730, China; Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
28
|
Wooff Y, Man SM, Aggio-Bruce R, Natoli R, Fernando N. IL-1 Family Members Mediate Cell Death, Inflammation and Angiogenesis in Retinal Degenerative Diseases. Front Immunol 2019; 10:1618. [PMID: 31379825 PMCID: PMC6646526 DOI: 10.3389/fimmu.2019.01618] [Citation(s) in RCA: 170] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 06/28/2019] [Indexed: 12/22/2022] Open
Abstract
Inflammation underpins and contributes to the pathogenesis of many retinal degenerative diseases. The recruitment and activation of both resident microglia and recruited macrophages, as well as the production of cytokines, are key contributing factors for progressive cell death in these diseases. In particular, the interleukin 1 (IL-1) family consisting of both pro- and anti-inflammatory cytokines has been shown to be pivotal in the mediation of innate immunity and contribute directly to a number of retinal degenerations, including Age-Related Macular Degeneration (AMD), diabetic retinopathy, retinitis pigmentosa, glaucoma, and retinopathy of prematurity (ROP). In this review, we will discuss the role of IL-1 family members and inflammasome signaling in retinal degenerative diseases, piecing together their contribution to retinal disease pathology, and identifying areas of research expansion required to further elucidate their function in the retina.
Collapse
Affiliation(s)
- Yvette Wooff
- The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia.,ANU Medical School, The Australian National University, Canberra, ACT, Australia
| | - Si Ming Man
- The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Riemke Aggio-Bruce
- The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Riccardo Natoli
- The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia.,ANU Medical School, The Australian National University, Canberra, ACT, Australia
| | - Nilisha Fernando
- The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| |
Collapse
|
29
|
Ristagno G, Fumagalli F, Bottazzi B, Mantovani A, Olivari D, Novelli D, Latini R. Pentraxin 3 in Cardiovascular Disease. Front Immunol 2019; 10:823. [PMID: 31057548 PMCID: PMC6481278 DOI: 10.3389/fimmu.2019.00823] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 03/28/2019] [Indexed: 12/30/2022] Open
Abstract
The long pentraxin PTX3 is a member of the pentraxin family produced locally by stromal and myeloid cells in response to proinflammatory signals and microbial moieties. The prototype of the pentraxin family is C reactive protein (CRP), a widely-used biomarker in human pathologies with an inflammatory or infectious origin. Data so far describe PTX3 as a multifunctional protein acting as a functional ancestor of antibodies and playing a regulatory role in inflammation. Cardiovascular disease (CVD) is a leading cause of mortality worldwide, and inflammation is crucial in promoting it. Data from animal models indicate that PTX3 can have cardioprotective and atheroprotective roles regulating inflammation. PTX3 has been investigated in several clinical settings as possible biomarker of CVD. Data collected so far indicate that PTX3 plasma levels rise rapidly in acute myocardial infarction, heart failure and cardiac arrest, reflecting the extent of tissue damage and predicting the risk of mortality.
Collapse
Affiliation(s)
- Giuseppe Ristagno
- Department of Cardiovascular Research, Mario Negri Institute for Pharmacological Research IRCCS, Milan, Italy
| | - Francesca Fumagalli
- Department of Cardiovascular Research, Mario Negri Institute for Pharmacological Research IRCCS, Milan, Italy
| | | | - Alberto Mantovani
- Humanitas Clinical and Research Center-IRCCS, Milan, Italy.,Humanitas University, Milan, Italy.,The William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Davide Olivari
- Department of Cardiovascular Research, Mario Negri Institute for Pharmacological Research IRCCS, Milan, Italy
| | - Deborah Novelli
- Department of Cardiovascular Research, Mario Negri Institute for Pharmacological Research IRCCS, Milan, Italy
| | - Roberto Latini
- Department of Cardiovascular Research, Mario Negri Institute for Pharmacological Research IRCCS, Milan, Italy
| |
Collapse
|
30
|
Luo Q, He X, Ning P, Zheng Y, Yang D, Xu Y, Shang Y, Gao Z. Admission Pentraxin-3 Level Predicts Severity of Community-Acquired Pneumonia Independently of Etiology. Proteomics Clin Appl 2019; 13:e1800117. [PMID: 30557448 DOI: 10.1002/prca.201800117] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 11/28/2018] [Indexed: 12/21/2022]
Abstract
PURPOSE Pentraxin-3 (PTX3) is a nonspecific marker of disease severity; however, PTX3 data from community-acquired pneumonia (CAP) patients are lacking. EXPERIMENTAL DESIGN An observational, prospective study of CAP patients was conducted in 2016. Plasma PTX3 levels are determined with quantitative ELISA. The primary endpoint is diagnosis of severe CAP (SCAP); the secondary endpoint is hospital mortality or discharge from the hospital. RESULTS Among 188 enrolled patients, 88 are diagnosed with SCAP, and 17 died during the hospital stay. Admission PTX3 levels are higher in patients with high CURB-65 (confusion, uremia, respiratory rate, blood pressure, age ≥65 years) or Pneumonia Severity Index (PSI) scores (p < 0.0001 for both) and are unaffected by etiology. PTX3 demonstrate good performance in predicting both SCAP in CAP patients (AUC = 0.847) and 30-day mortality of CAP patients (AUC = 0.796). PTX3 combined with CURB-65 improve prediction performance (p = 0.0379). Furthermore, multivariate Cox regression analysis confirm ≥33.52 ng mL-1 PTX3 as an independent predictor of 30-day survival. CONCLUSIONS AND CLINICAL RELEVANCE Admission levels of PTX3 are useful for predicting severity of CAP, independent of possible pathogens. PTX3 can improve prognostic accuracy of severity scores. Detection of PTX3 on admission might be useful for clinical judgment. TRIAL REGISTRATION ClinicalTrials.gov ID: NCT03093220, Date of registration: March 28, 2017 (retrospectively registered).
Collapse
Affiliation(s)
- Qiongzhen Luo
- Department of Respiratory and Critical Care Medicine, Peking University People's Hospital, Beijing, Xicheng, 100044, P. R. China
| | - Xinwei He
- Department of Internal Medicine, Xicheng District Zhanlanlu Hospital, Beijing, Xicheng, 100044, P. R. China
| | - Pu Ning
- Department of Respiratory and Critical Care Medicine, Peking University People's Hospital, Beijing, Xicheng, 100044, P. R. China
| | - Yali Zheng
- Department of Respiratory and Critical Care Medicine, Peking University People's Hospital, Beijing, Xicheng, 100044, P. R. China
| | - Donghong Yang
- Department of Respiratory and Critical Care Medicine, Peking University People's Hospital, Beijing, Xicheng, 100044, P. R. China
| | - Yu Xu
- Department of Respiratory and Critical Care Medicine, Peking University People's Hospital, Beijing, Xicheng, 100044, P. R. China
| | - Ying Shang
- Department of Respiratory and Critical Care Medicine, Peking University People's Hospital, Beijing, Xicheng, 100044, P. R. China
| | - Zhancheng Gao
- Department of Respiratory and Critical Care Medicine, Peking University People's Hospital, Beijing, Xicheng, 100044, P. R. China
| |
Collapse
|
31
|
Donato L, Scimone C, Nicocia G, D'Angelo R, Sidoti A. Role of oxidative stress in Retinitis pigmentosa: new involved pathways by an RNA-Seq analysis. Cell Cycle 2018; 18:84-104. [PMID: 30569795 DOI: 10.1080/15384101.2018.1558873] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Retinitis pigmentosa (RP) is a very heterogeneous inherited ocular disorder group characterized by progressive retinal disruption. Retinal pigment epithelium (RPE) degeneration, due to oxidative stress which arrests the metabolic support to photoreceptors, represents one of the principal causes of RP. Here, the role of oxidative stress in RP onset and progression was analyzed by a comparative whole transcriptome analysis of human RPE cells, treated with 100 µg/ml of oxLDL and untreated, at different time points. Experiment was thrice repeated and performed on Ion ProtonTM sequencing system. Data analysis, including low quality reads trimming and gene expression quantification, was realized by CLC Genomics Workbench software. The whole analysis highlighted 14 clustered "macro-pathways" and many sub-pathways, classified by selection of 5271 genes showing the highest alteration of expression. Among them, 23 genes were already known to be RP causative ones (15 over-expressed and 8 down-expressed), and their enrichment and intersection analyses highlighted new 77 candidate related genes (49 over-expressed and 28 down-expressed). A final filtering analysis then highlighted 29 proposed candidate genes. This data suggests that many new genes, not yet associated with RP, could influence its etiopathogenesis.
Collapse
Affiliation(s)
- Luigi Donato
- a Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Division of Medical Biotechnologies and Preventive Medicine , University of Messina , Messina , Italy.,b Department of Cutting-Edge Medicine and Therapies, Biomolecular Strategies and Neuroscience, Section of Applied Neuroscience, Molecular Genetics and Predictive Medicine , I.E.ME.S.T. ., Palermo , Italy
| | - Concetta Scimone
- a Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Division of Medical Biotechnologies and Preventive Medicine , University of Messina , Messina , Italy.,b Department of Cutting-Edge Medicine and Therapies, Biomolecular Strategies and Neuroscience, Section of Applied Neuroscience, Molecular Genetics and Predictive Medicine , I.E.ME.S.T. ., Palermo , Italy
| | - Giacomo Nicocia
- c Department of Clinical and Experimental Medicine , University of Messina , Messina , Italy
| | - Rosalia D'Angelo
- a Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Division of Medical Biotechnologies and Preventive Medicine , University of Messina , Messina , Italy.,b Department of Cutting-Edge Medicine and Therapies, Biomolecular Strategies and Neuroscience, Section of Applied Neuroscience, Molecular Genetics and Predictive Medicine , I.E.ME.S.T. ., Palermo , Italy
| | - Antonina Sidoti
- a Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Division of Medical Biotechnologies and Preventive Medicine , University of Messina , Messina , Italy.,b Department of Cutting-Edge Medicine and Therapies, Biomolecular Strategies and Neuroscience, Section of Applied Neuroscience, Molecular Genetics and Predictive Medicine , I.E.ME.S.T. ., Palermo , Italy
| |
Collapse
|
32
|
Ma YJ, Garred P. Pentraxins in Complement Activation and Regulation. Front Immunol 2018; 9:3046. [PMID: 30619374 PMCID: PMC6305747 DOI: 10.3389/fimmu.2018.03046] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 12/10/2018] [Indexed: 01/10/2023] Open
Abstract
The complement is the first line of immune defense system involved in elimination of invading pathogens and dying host cells. Its activation is mainly triggered by immune complexes or pattern recognition molecules (PRMs) upon recognition against non-self or altered self-cells, such as C1q, collectins, ficolins, and properdin. Recent findings have interestingly shown that the pentraxins (C-reactive protein, CRP; serum-amyloid P component, SAP; long pentraxin 3, PTX3) are involved in complement activation and amplification via communication with complement initiation PRMs, but also complement regulation via recruitment of complement regulators, for instance C4b binding protein (C4BP) and factor H (fH). This review addresses the potential roles of the pentraxins in the complement system during infection and inflammation, and emphasizes the underlining implications of the pentraxins in the context of complement activation and regulation both under physiological and pathological conditions.
Collapse
Affiliation(s)
- Ying Jie Ma
- The Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Rigshospitalet, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Peter Garred
- The Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Rigshospitalet, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
33
|
Garlanda C, Bottazzi B, Magrini E, Inforzato A, Mantovani A. PTX3, a Humoral Pattern Recognition Molecule, in Innate Immunity, Tissue Repair, and Cancer. Physiol Rev 2018; 98:623-639. [PMID: 29412047 DOI: 10.1152/physrev.00016.2017] [Citation(s) in RCA: 142] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Innate immunity includes a cellular and a humoral arm. PTX3 is a fluid-phase pattern recognition molecule conserved in evolution which acts as a key component of humoral innate immunity in infections of fungal, bacterial, and viral origin. PTX3 binds conserved microbial structures and self-components under conditions of inflammation and activates effector functions (complement, phagocytosis). Moreover, it has a complex regulatory role in inflammation, such as ischemia/reperfusion injury and cancer-related inflammation, as well as in extracellular matrix organization and remodeling, with profound implications in physiology and pathology. Finally, PTX3 acts as an extrinsic oncosuppressor gene by taming tumor-promoting inflammation in murine and selected human tumors. Thus evidence suggests that PTX3 is a key homeostatic component at the crossroad of innate immunity, inflammation, tissue repair, and cancer. Dissecting the complexity of PTX3 pathophysiology and human genetics paves the way to diagnostic and therapeutic exploitation.
Collapse
Affiliation(s)
- Cecilia Garlanda
- Humanitas Clinical and Research Center, Rozzano, Milan , Italy ; Humanitas University, Rozzano, Milan , Italy ; Department of Medical Biotechnologies and Translational Medicine, University of Milan , Milan , Italy ; and The William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Barbara Bottazzi
- Humanitas Clinical and Research Center, Rozzano, Milan , Italy ; Humanitas University, Rozzano, Milan , Italy ; Department of Medical Biotechnologies and Translational Medicine, University of Milan , Milan , Italy ; and The William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Elena Magrini
- Humanitas Clinical and Research Center, Rozzano, Milan , Italy ; Humanitas University, Rozzano, Milan , Italy ; Department of Medical Biotechnologies and Translational Medicine, University of Milan , Milan , Italy ; and The William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Antonio Inforzato
- Humanitas Clinical and Research Center, Rozzano, Milan , Italy ; Humanitas University, Rozzano, Milan , Italy ; Department of Medical Biotechnologies and Translational Medicine, University of Milan , Milan , Italy ; and The William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Alberto Mantovani
- Humanitas Clinical and Research Center, Rozzano, Milan , Italy ; Humanitas University, Rozzano, Milan , Italy ; Department of Medical Biotechnologies and Translational Medicine, University of Milan , Milan , Italy ; and The William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
34
|
Ebrahimi KB, Cano M, Rhee J, Datta S, Wang L, Handa JT. Oxidative Stress Induces an Interactive Decline in Wnt and Nrf2 Signaling in Degenerating Retinal Pigment Epithelium. Antioxid Redox Signal 2018; 29:389-407. [PMID: 29186981 PMCID: PMC6025703 DOI: 10.1089/ars.2017.7084] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
AIMS Cells have evolved a highly sophisticated web of cytoprotective systems to neutralize unwanted oxidative stress, but are challenged by unique modern day stresses such as cigarette smoking and ingestion of a high-fat diet (HFD). Age-related disease, such as age-related macular degeneration (AMD), the most common cause of blindness among the elderly in Western societies, develops in part, when oxidative stress overwhelms cytoprotective systems to injure tissue. Since most studies focus on the protection by a single protective system, the aim of this study was to investigate the impact of more than one cytoprotective system against oxidative stress. RESULTS Wingless (Wnt) and nuclear factor-erythroid 2-related factor 2 (Nrf2), two fundamental signaling systems that are vital to cell survival, decline after mice are exposed to chronic cigarette smoke and HFD, two established AMD risk factors, in a bidirectional feedback loop through phosphorylated glycogen synthase kinase 3 beta. Decreased Wnt and Nrf2 signaling leads to retinal pigment epithelial dysfunction and apoptosis, and a phenotype that is strikingly similar to geographic atrophy (GA), an advanced form of AMD with no effective treatment. INNOVATION This study is the first to show that chronic oxidative stress from common modern day environmental exposures reduces two fundamental and vital cytoprotective networks in a bidirectional feedback loop, and their decline leads to advanced disease phenotype. CONCLUSION Our data offer new insights into how combined modern oxidative stresses of cigarette smoking and HFD contribute to GA through an interactive decline in Wnt and Nrf2 signaling. Antioxid. Redox Signal. 29, 389-407.
Collapse
Affiliation(s)
- Katayoon B Ebrahimi
- Wilmer Eye Institute , Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Marisol Cano
- Wilmer Eye Institute , Johns Hopkins School of Medicine, Baltimore, Maryland
| | - John Rhee
- Wilmer Eye Institute , Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Sayantan Datta
- Wilmer Eye Institute , Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Lei Wang
- Wilmer Eye Institute , Johns Hopkins School of Medicine, Baltimore, Maryland
| | - James T Handa
- Wilmer Eye Institute , Johns Hopkins School of Medicine, Baltimore, Maryland
| |
Collapse
|
35
|
C-reactive protein and pentraxin-3 binding of factor H-like protein 1 differs from complement factor H: implications for retinal inflammation. Sci Rep 2018; 8:1643. [PMID: 29374201 PMCID: PMC5786067 DOI: 10.1038/s41598-017-18395-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 12/11/2017] [Indexed: 12/31/2022] Open
Abstract
Retinal inflammation plays a key role in the progression of age-related macular degeneration (AMD), a condition that leads to loss of central vision. The deposition of the acute phase pentraxin C-reactive protein (CRP) in the macula activates the complement system, thereby contributing to dysregulated inflammation. The complement protein factor H (FH) can bind CRP and down-regulate an inflammatory response. However, it is not known whether a truncated form of FH, called factor H-like protein 1 (FHL-1), which plays a significant regulatory role in the eye, also interacts with CRP. Here, we compare the binding properties of FHL-1 and FH to both CRP and the related protein pentraxin-3 (PTX3). We find that, unlike FH, FHL-1 can bind pro-inflammatory monomeric CRP (mCRP) as well as the circulating pentameric form. Furthermore, the four-amino acid C-terminal tail of FHL-1 (not present in FH) plays a role in mediating its binding to mCRP. PTX3 was found to be present in the macula of donor eyes and the AMD-associated Y402H polymorphism altered the binding of FHL-1 to PTX3. Our findings reveal that the binding characteristics of FHL-1 differ from those of FH, likely underpinning independent immune regulatory functions in the context of the human retina.
Collapse
|
36
|
Kosmidou C, Efstathiou NE, Hoang MV, Notomi S, Konstantinou EK, Hirano M, Takahashi K, Maidana DE, Tsoka P, Young L, Gragoudas ES, Olsen TW, Morizane Y, Miller JW, Vavvas DG. Issues with the Specificity of Immunological Reagents for NLRP3: Implications for Age-related Macular Degeneration. Sci Rep 2018; 8:461. [PMID: 29323137 PMCID: PMC5764999 DOI: 10.1038/s41598-017-17634-1] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 11/28/2017] [Indexed: 11/17/2022] Open
Abstract
Contradictory data have been presented regarding the implication of the NACHT, LRR and PYD domains-containing protein 3 (NLRP3) inflammasome in age-related macular degeneration (AMD), the leading cause of vision loss in the Western world. Recognizing that antibody specificity may explain this discrepancy and in line with recent National Institutes of Health (NIH) guidelines requiring authentication of key biological resources, the specificity of anti-NLRP3 antibodies was assessed to elucidate whether non-immune RPE cells express NLRP3. Using validated resources, NLRP3 was not detected in human primary or human established RPE cell lines under multiple inflammasome-priming conditions, including purported NLRP3 stimuli in RPE such as DICER1 deletion and Alu RNA transfection. Furthermore, NLRP3 was below detection limits in ex vivo macular RPE from AMD patients, as well as in human induced pluripotent stem cell (hiPSC)-derived RPE from patients with overactive NLRP3 syndrome (Chronic infantile neurologic cutaneous and articulate, CINCA syndrome). Evidence presented in this study provides new data regarding the interpretation of published results reporting NLRP3 expression and upregulation in RPE and addresses the role that this inflammasome plays in AMD pathogenesis.
Collapse
Affiliation(s)
- Cassandra Kosmidou
- Department of Ophthalmology, Retina Service, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, Massachusetts, 02114, USA
| | - Nikolaos E Efstathiou
- Department of Ophthalmology, Retina Service, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, Massachusetts, 02114, USA
| | - Mien V Hoang
- Department of Ophthalmology, Retina Service, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, Massachusetts, 02114, USA
| | - Shoji Notomi
- Department of Ophthalmology, Retina Service, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, Massachusetts, 02114, USA
| | - Eleni K Konstantinou
- Department of Ophthalmology, Retina Service, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, Massachusetts, 02114, USA
| | - Masayuki Hirano
- Department of Ophthalmology, Okayama University Graduate School of Medicine, Okayama, 700-8558, Japan
| | - Kosuke Takahashi
- Department of Ophthalmology, Okayama University Graduate School of Medicine, Okayama, 700-8558, Japan
| | - Daniel E Maidana
- Department of Ophthalmology, Retina Service, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, Massachusetts, 02114, USA
| | - Pavlina Tsoka
- Department of Ophthalmology, Retina Service, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, Massachusetts, 02114, USA
| | - Lucy Young
- Department of Ophthalmology, Retina Service, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, Massachusetts, 02114, USA
| | - Evangelos S Gragoudas
- Department of Ophthalmology, Retina Service, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, Massachusetts, 02114, USA
| | - Timothy W Olsen
- Department of Ophthalmology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Yuki Morizane
- Department of Ophthalmology, Okayama University Graduate School of Medicine, Okayama, 700-8558, Japan
| | - Joan W Miller
- Department of Ophthalmology, Retina Service, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, Massachusetts, 02114, USA
| | - Demetrios G Vavvas
- Department of Ophthalmology, Retina Service, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, Massachusetts, 02114, USA.
| |
Collapse
|
37
|
Ischemic Retinopathies: Oxidative Stress and Inflammation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:3940241. [PMID: 29410732 PMCID: PMC5749295 DOI: 10.1155/2017/3940241] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Accepted: 11/20/2017] [Indexed: 12/15/2022]
Abstract
Ischemic retinopathies (IRs), such as retinopathy of prematurity (ROP), diabetic retinopathy (DR), and (in many cases) age-related macular degeneration (AMD), are ocular disorders characterized by an initial phase of microvascular changes that results in ischemia, followed by a second phase of abnormal neovascularization that may culminate into retinal detachment and blindness. IRs are complex retinal conditions in which several factors play a key role during the development of the different pathological stages of the disease. Increasing evidence reveals that oxidative stress and inflammatory processes are important contributors to the pathogenesis of IRs. Despite the beneficial effects of the photocoagulation and anti-VEGF therapy during neovascularization phase, the need to identify novel targets to prevent initial phases of these ocular pathologies is still needed. In this review, we provide an update on the involvement of oxidative stress and inflammation in the progression of IRs and address some therapeutic interventions by using antioxidants and anti-inflammatory agents.
Collapse
|
38
|
The impact of oxidative stress and inflammation on RPE degeneration in non-neovascular AMD. Prog Retin Eye Res 2017; 60:201-218. [PMID: 28336424 DOI: 10.1016/j.preteyeres.2017.03.002] [Citation(s) in RCA: 551] [Impact Index Per Article: 68.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 03/13/2017] [Accepted: 03/14/2017] [Indexed: 02/07/2023]
Abstract
The retinal pigment epithelium (RPE) is a highly specialized, unique epithelial cell that interacts with photoreceptors on its apical side and with Bruch's membrane and the choriocapillaris on its basal side. Due to vital functions that keep photoreceptors healthy, the RPE is essential for maintaining vision. With aging and the accumulated effects of environmental stresses, the RPE can become dysfunctional and die. This degeneration plays a central role in age-related macular degeneration (AMD) pathobiology, the leading cause of blindness among the elderly in western societies. Oxidative stress and inflammation have both physiological and potentially pathological roles in RPE degeneration. Given the central role of the RPE, this review will focus on the impact of oxidative stress and inflammation on the RPE with AMD pathobiology. Physiological sources of oxidative stress as well as unique sources from photo-oxidative stress, the phagocytosis of photoreceptor outer segments, and modifiable factors such as cigarette smoking and high fat diet ingestion that can convert oxidative stress into a pathological role, and the negative impact of impairing the cytoprotective roles of mitochondrial dynamics and the Nrf2 signaling system on RPE health in AMD will be discussed. Likewise, the response by the innate immune system to an inciting trigger, and the potential role of local RPE production of inflammation, as well as a potential role for damage by inflammation with chronicity if the inciting trigger is not neutralized, will be debated.
Collapse
|