1
|
Vastrad B, Vastrad C. Screening and identification of key biomarkers associated with endometriosis using bioinformatics and next-generation sequencing data analysis. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2024; 25:116. [DOI: 10.1186/s43042-024-00572-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 08/23/2024] [Indexed: 01/04/2025] Open
Abstract
Abstract
Background
Endometriosis is a common cause of endometrial-type mucosa outside the uterine cavity with symptoms such as painful periods, chronic pelvic pain, pain with intercourse and infertility. However, the early diagnosis of endometriosis is still restricted. The purpose of this investigation is to identify and validate the key biomarkers of endometriosis.
Methods
Next-generation sequencing dataset GSE243039 was obtained from the Gene Expression Omnibus database, and differentially expressed genes (DEGs) between endometriosis and normal control samples were identified. After screening of DEGs, gene ontology (GO) and REACTOME pathway enrichment analyses were performed. Furthermore, a protein–protein interaction (PPI) network was constructed and modules were analyzed using the Human Integrated Protein–Protein Interaction rEference database and Cytoscape software, and hub genes were identified. Subsequently, a network between miRNAs and hub genes, and network between TFs and hub genes were constructed using the miRNet and NetworkAnalyst tool, and possible key miRNAs and TFs were predicted. Finally, receiver operating characteristic curve analysis was used to validate the hub genes.
Results
A total of 958 DEGs, including 479 upregulated genes and 479 downregulated genes, were screened between endometriosis and normal control samples. GO and REACTOME pathway enrichment analyses of the 958 DEGs showed that they were mainly involved in multicellular organismal process, developmental process, signaling by GPCR and muscle contraction. Further analysis of the PPI network and modules identified 10 hub genes, including vcam1, snca, prkcb, adrb2, foxq1, mdfi, actbl2, prkd1, dapk1 and actc1. Possible target miRNAs, including hsa-mir-3143 and hsa-mir-2110, and target TFs, including tcf3 (transcription factor 3) and clock (clock circadian regulator), were predicted by constructing a miRNA-hub gene regulatory network and TF-hub gene regulatory network.
Conclusions
This investigation used bioinformatics techniques to explore the potential and novel biomarkers. These biomarkers might provide new ideas and methods for the early diagnosis, treatment and monitoring of endometriosis.
Collapse
|
2
|
Prieto-López L, Pereiro X, Vecino E. The mechanics of the retina: Müller glia role on retinal extracellular matrix and modelling. Front Med (Lausanne) 2024; 11:1393057. [PMID: 39296899 PMCID: PMC11410058 DOI: 10.3389/fmed.2024.1393057] [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: 03/04/2024] [Accepted: 08/13/2024] [Indexed: 09/21/2024] Open
Abstract
The retina is a highly heterogeneous tissue, both cell-wise but also regarding its extracellular matrix (ECM). The stiffness of the ECM is pivotal in retinal development and maturation and has also been associated with the onset and/or progression of numerous retinal pathologies, such as glaucoma, proliferative vitreoretinopathy (PVR), age-related macular degeneration (AMD), epiretinal membrane (ERM) formation or uveitis. Nonetheless, much remains unknown about the biomechanical milieu of the retina, and specifically the role that Müller glia play as principal mechanosensors and major producers of ECM constituents. So far, new approaches need to be developed to further the knowledge in the field of retinal mechanobiology for ECM-target applications to arise. In this review, we focus on the involvement of Müller glia in shaping and altering the retinal ECM under both physiological and pathological conditions and look into various biomaterial options to more accurately replicate the impact of matrix stiffness in vitro.
Collapse
Affiliation(s)
- Laura Prieto-López
- Experimental Ophthalmo-Biology Group, Department of Cell Biology and Histology, University of Basque Country UPV/EHU, Leioa, Spain
| | - Xandra Pereiro
- Experimental Ophthalmo-Biology Group, Department of Cell Biology and Histology, University of Basque Country UPV/EHU, Leioa, Spain
- Begiker-Ophthalmology Research Group, BioCruces Health Research Institute, Cruces Hospital, Barakaldo, Spain
| | - Elena Vecino
- Experimental Ophthalmo-Biology Group, Department of Cell Biology and Histology, University of Basque Country UPV/EHU, Leioa, Spain
- Begiker-Ophthalmology Research Group, BioCruces Health Research Institute, Cruces Hospital, Barakaldo, Spain
| |
Collapse
|
3
|
Zhang Y, Zhang Z, Mo Y, Zhang Y, Yuan J, Zhang Q. MMP-3 mediates copper oxide nanoparticle-induced pulmonary inflammation and fibrosis. J Nanobiotechnology 2024; 22:428. [PMID: 39030581 PMCID: PMC11264740 DOI: 10.1186/s12951-024-02707-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 07/05/2024] [Indexed: 07/21/2024] Open
Abstract
BACKGROUND The increasing production and usage of copper oxide nanoparticles (Nano-CuO) raise human health concerns. Previous studies have demonstrated that exposure to Nano-CuO could induce lung inflammation, injury, and fibrosis. However, the potential underlying mechanisms are still unclear. Here, we proposed that matrix metalloproteinase-3 (MMP-3) might play an important role in Nano-CuO-induced lung inflammation, injury, and fibrosis. RESULTS Exposure of mice to Nano-CuO caused acute lung inflammation and injury in a dose-dependent manner, which was reflected by increased total cell number, neutrophil count, macrophage count, lactate dehydrogenase (LDH) activity, and CXCL1/KC level in bronchoalveolar lavage fluid (BALF) obtained on day 3 post-exposure. The time-response study showed that Nano-CuO-induced acute lung inflammation and injury appeared as early as day 1 after exposure, peaked on day 3, and ameliorated over time. However, even on day 42 post-exposure, the LDH activity and macrophage count were still higher than those in the control group, suggesting that Nano-CuO caused chronic lung inflammation. The Nano-CuO-induced pulmonary inflammation was further confirmed by H&E staining of lung sections. Trichrome staining showed that Nano-CuO exposure caused pulmonary fibrosis from day 14 to day 42 post-exposure with an increasing tendency over time. Increased hydroxyproline content and expression levels of fibrosis-associated proteins in mouse lungs were also observed. In addition, Nano-CuO exposure induced MMP-3 overexpression and increased MMP-3 secretion in mouse lungs. Knocking down MMP-3 in mouse lungs significantly attenuated Nano-CuO-induced acute and chronic lung inflammation and fibrosis. Moreover, Nano-CuO exposure caused sustained production of cleaved osteopontin (OPN) in mouse lungs, which was also significantly decreased by knocking down MMP-3. CONCLUSIONS Our results demonstrated that short-term Nano-CuO exposure caused acute lung inflammation and injury, while long-term exposure induced chronic pulmonary inflammation and fibrosis. Knocking down MMP-3 significantly ameliorated Nano-CuO-induced pulmonary inflammation, injury, and fibrosis, and also attenuated Nano-CuO-induced cleaved OPN level. Our study suggests that MMP-3 may play important roles in Nano-CuO-induced pulmonary inflammation and fibrosis via cleavage of OPN and may provide a further understanding of the mechanisms underlying Nano-CuO-induced pulmonary toxicity.
Collapse
Affiliation(s)
- Yuanbao Zhang
- Department of Epidemiology and Population Health, School of Public Health and Information Sciences, University of Louisville, 485 E. Gray Street, Louisville, KY, 40202, USA
- Beijing Key Laboratory of Occupational Safety and Health, Institute of Urban Safety and Environmental Science, Beijing Academy of Science and Technology, Beijing, 100054, China
| | - Zhenyu Zhang
- Department of Emergency, Xiang'An Hospital of Xiamen University, Xiamen, 361104, Fujian, China
| | - Yiqun Mo
- Department of Epidemiology and Population Health, School of Public Health and Information Sciences, University of Louisville, 485 E. Gray Street, Louisville, KY, 40202, USA
| | - Yue Zhang
- Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Jiali Yuan
- Department of Epidemiology and Population Health, School of Public Health and Information Sciences, University of Louisville, 485 E. Gray Street, Louisville, KY, 40202, USA
| | - Qunwei Zhang
- Department of Epidemiology and Population Health, School of Public Health and Information Sciences, University of Louisville, 485 E. Gray Street, Louisville, KY, 40202, USA.
| |
Collapse
|
4
|
Torilis japonica Extract Suppresses the Induction of Atopic Inflammation. Int J Mol Sci 2023; 24:ijms24032102. [PMID: 36768424 PMCID: PMC9916524 DOI: 10.3390/ijms24032102] [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: 12/26/2022] [Revised: 01/12/2023] [Accepted: 01/17/2023] [Indexed: 01/21/2023] Open
Abstract
As one of the major intractable allergic disorders, atopic inflammation is commonly accompanied by itching, dry skin, and inflammation. Atopic inflammation deteriorates the quality of life and has no fundamental cure, so it is crucial to urgently explore and develop natural resources for long-term treatment without any side effects. This study aimed to verify Torilis japonica extract (TJE)'s relieving effect and mechanism against atopic inflammation using skin cells and skin equivalent models, as well as to investigate torilin's effect (obtained from TJE) and other unknown components as marker compounds. Torilin concentration was verified in TJE using high-performance liquid chromatography and analyzed the unknown components using nuclear magnetic resonance spectroscopy. Furthermore, TJE's cytotoxicity, regenerative effect, and cell cycle regulation effects were confirmed using skin cells with atopic inflammation (human dermal fibroblasts and HaCaT keratinocytes) by using TNF-α and IFN-γ treatments. Consequently, TJE was demonstrated to regulate TARC and CTACK expressions as chemokines and those of interleukin-4, -5, and -13 as cytokines related to atopic inflammation. TJE was further confirmed to affect the matrix metalloproteinase-1, -2, and -9 expressions, which are essential in skin damage. Lastly, this study confirmed TJE's relieving effect against atopic inflammation through a 3D skin model and RhCE model using human dermal fibroblasts and HaCaT keratinocytes. These findings on atopic inflammation verified torilin's relieving effects and TJE's other components.
Collapse
|
5
|
Biasella F, Plössl K, Baird PN, Weber BHF. The extracellular microenvironment in immune dysregulation and inflammation in retinal disorders. Front Immunol 2023; 14:1147037. [PMID: 36936905 PMCID: PMC10014728 DOI: 10.3389/fimmu.2023.1147037] [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: 01/18/2023] [Accepted: 02/15/2023] [Indexed: 03/05/2023] Open
Abstract
Inherited retinal dystrophies (IRDs) as well as genetically complex retinal phenotypes represent a heterogenous group of ocular diseases, both on account of their phenotypic and genotypic characteristics. Therefore, overlaps in clinical features often complicate or even impede their correct clinical diagnosis. Deciphering the molecular basis of retinal diseases has not only aided in their disease classification but also helped in our understanding of how different molecular pathologies may share common pathomechanisms. In particular, these relate to dysregulation of two key processes that contribute to cellular integrity, namely extracellular matrix (ECM) homeostasis and inflammation. Pathological changes in the ECM of Bruch's membrane have been described in both monogenic IRDs, such as Sorsby fundus dystrophy (SFD) and Doyne honeycomb retinal dystrophy (DHRD), as well as in the genetically complex age-related macular degeneration (AMD) or diabetic retinopathy (DR). Additionally, complement system dysfunction and distorted immune regulation may also represent a common connection between some IRDs and complex retinal degenerations. Through highlighting such overlaps in molecular pathology, this review aims to illuminate how inflammatory processes and ECM homeostasis are linked in the healthy retina and how their interplay may be disturbed in aging as well as in disease.
Collapse
Affiliation(s)
- Fabiola Biasella
- Institute of Human Genetics, University of Regensburg, Regensburg, Germany
| | - Karolina Plössl
- Institute of Human Genetics, University of Regensburg, Regensburg, Germany
| | - Paul N. Baird
- Institute of Human Genetics, University of Regensburg, Regensburg, Germany
- Department of Surgery, Ophthalmology, University of Melbourne, Melbourne, VIC, Australia
- *Correspondence: Paul N. Baird, ; Bernhard H. F. Weber,
| | - Bernhard H. F. Weber
- Institute of Human Genetics, University of Regensburg, Regensburg, Germany
- Institute of Clinical Human Genetics, University Hospital Regensburg, Regensburg, Germany
- *Correspondence: Paul N. Baird, ; Bernhard H. F. Weber,
| |
Collapse
|
6
|
Xiao X, Liu Z, Su G, Liu H, Yin W, Guan Y, Jing S, Du L, Li F, Li N, Yang P. A novel uveitis model induced by lipopolysaccharide in zebrafish. Front Immunol 2022; 13:1042849. [PMID: 36532084 PMCID: PMC9751191 DOI: 10.3389/fimmu.2022.1042849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 11/18/2022] [Indexed: 12/03/2022] Open
Abstract
Objective Endotoxin-induced uveitis (EIU) is an important tool for human uveitis study. This study was designed to develop a novel EIU model in zebrafish. Methods An EIU model in zebrafish was induced by intravitreal lipopolysaccharide (LPS) injection and was assessed dynamically. Optical coherence tomography (OCT) was used to assess infiltrating cells in the vitreous body. The histological changes wereevaluated using HE staining and immune cells were measured by immunofluorescence. The retinal RNA Sequencing (RNA-Seq) was used to explore the transcriptional changes during inflammation. RNA-Seq data were analyzed using time-course sequencing data analysis (TCseq), ClueGO plugin in Cytoscape, and Gene Set Enrichment Analysis (GSEA) software. Flow cytometry and retinal flat mounts were used to dynamically quantify the immune cells. Results EIU was successfully induced in zebrafish following intravitreal LPS injection. Inflammation appeared at 4 hours post injection (hpi), reached its peak at 24 hpi, and then resolved at 72 hpi. Immunofluorescence confirmed that massive influx ofneutrophils into the iris and vitreous body, and activation of microglia as evidenced by ameboid-shaped appearance in the retina. Retinal RNA-seq during the EIU course identified four gene clusters with distinct expression characteristics related to Toll-likereceptor signaling pathway, cytokine-cytokine receptor interaction, NOD-like receptor signaling pathway, and extracellular matrix (ECM)-receptor interaction, respectively. Prednisone immersion inhibited the inflammatory response of EIU in zebrafish, whichwas confirmed by decreased neutrophils detected in flow cytometry and retinal flat mounts. Conclusions We developed a novel EIU model in zebrafish, which may be particularly useful for gene-editing and high-throughput screening of new drugs for the prevention and treatment of uveitis.
Collapse
Affiliation(s)
- Xiao Xiao
- Henan International Joint Research Laboratory for Ocular Immunology and Retinal Injury Repair, Henan Province Eye Hospital, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhangluxi Liu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology and Chongqing Eye Institute, Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing, China
| | - Guannan Su
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology and Chongqing Eye Institute, Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing, China
| | - Huan Liu
- Henan International Joint Research Laboratory for Ocular Immunology and Retinal Injury Repair, Henan Province Eye Hospital, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wenhui Yin
- Henan International Joint Research Laboratory for Ocular Immunology and Retinal Injury Repair, Henan Province Eye Hospital, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yuxuan Guan
- Henan International Joint Research Laboratory for Ocular Immunology and Retinal Injury Repair, Henan Province Eye Hospital, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shixiang Jing
- Henan International Joint Research Laboratory for Ocular Immunology and Retinal Injury Repair, Henan Province Eye Hospital, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Liping Du
- Henan International Joint Research Laboratory for Ocular Immunology and Retinal Injury Repair, Henan Province Eye Hospital, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Fuzhen Li
- Henan International Joint Research Laboratory for Ocular Immunology and Retinal Injury Repair, Henan Province Eye Hospital, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Na Li
- Henan International Joint Research Laboratory for Ocular Immunology and Retinal Injury Repair, Henan Province Eye Hospital, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Peizeng Yang
- Henan International Joint Research Laboratory for Ocular Immunology and Retinal Injury Repair, Henan Province Eye Hospital, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China,The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology and Chongqing Eye Institute, Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing, China,*Correspondence: Peizeng Yang,
| |
Collapse
|
7
|
Jones TK, Reilly JP, Anderson BJ, Miano TA, Dunn TG, Weisman AR, Agyekum R, Feng R, Ittner CA, Shashaty MG, Meyer NJ. Elevated Plasma Levels of Matrix Metalloproteinase-3 and Tissue-Inhibitor of Matrix Metalloproteinases-1 Associate With Organ Dysfunction and Mortality in Sepsis. Shock 2022; 57:41-47. [PMID: 34265829 PMCID: PMC8663538 DOI: 10.1097/shk.0000000000001833] [Citation(s) in RCA: 16] [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: 04/10/2021] [Accepted: 06/23/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND Matrix Metalloproteinases (MMP) respond to tissue damage during sepsis. Higher plasma concentrations of MMPs and the tissue-inhibitor of matrix metalloproteinases (TIMP) have been reported in sepsis compared with healthy controls. The objective of this study was to examine if plasma levels of MMP-3, MMP-9, and TIMP-1 associate with mortality and organ dysfunction during sepsis. METHODS We conducted a prospective cohort study of critically ill patients with sepsis adjudicated per Sepsis-3 criteria at a tertiary academic medical center. We measured plasma concentrations of MMP-3, MMP-9, and TIMP-1 on intensive care unit admission. We phenotyped the subjects for shock, acute respiratory distress syndrome (ARDS), acute kidney injury (AKI), and mortality at 30 days. We used logistic regression to test the associations between the MMPs and TIMP-1 with shock, ARDS, AKI, and mortality. RESULTS Higher plasma TIMP-1 levels were associated with shock (odds ratio [OR] 1.51 per log increase [95% CI 1.25, 1.83]), ARDS (OR 1.24 [95% CI 1.05, 1.46]), AKI (OR 1.18 [95% CI 1.01, 1.38]), and mortality (OR 1.20 [95% CI 1.05, 1.46]. Higher plasma MMP-3 concentrations were associated with shock (OR 1.40 [95% CI 1.12, 1.75]) and mortality (OR 1.24 [95% CI 1.03, 1.48]) whereas MMP-9 levels were not associated with outcomes. Higher plasma TIMP-1 to MMP-3 ratios were associated with shock (OR 1.41 [95% CI 1.15, 1.72], P = 0.02). CONCLUSION Elevated plasma concentrations of TIMP-1 associate with organ dysfunction and mortality in sepsis. Higher plasma levels of MMP-3 associate with shock and mortality. Plasma MMP and TIMP-1 may warrant further investigation as emerging sepsis theragnostic biomarkers.
Collapse
Affiliation(s)
- Tiffanie K. Jones
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - John P. Reilly
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Brian J. Anderson
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Todd A. Miano
- Division of Epidemiology, Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Thomas G. Dunn
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ariel R. Weisman
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Roseline Agyekum
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Rui Feng
- Division of Biostatistics, Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Caroline A.G. Ittner
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Michael G.S. Shashaty
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Nuala J. Meyer
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
8
|
Hu DN, Zhang R, Yao S, Iacob CE, Yang WE, Rosen R, Yang SF. Cultured Human Uveal Melanocytes Express/secrete CXCL1 and CXCL2 Constitutively and Increased by Lipopolysaccharide via Activation of Toll-like Receptor 4. Curr Eye Res 2021; 46:1681-1694. [PMID: 33979551 DOI: 10.1080/02713683.2021.1929326] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 05/01/2021] [Accepted: 05/03/2021] [Indexed: 02/07/2023]
Abstract
Purpose: Lipopolysaccharide (LPS) can activate Toll-like receptor 4 (TLR4) and increase the expression of CXCL1 and CXCL2, the potent neutrophils chemoattractants, in various cell types. These effects have not been previously reported in the uveal melanocytes. This study was designed to investigate the effects of LPS on the activation of TLR4 and expression of CXCL1/CXCL2 in cultured human uveal melanocytes and the relevant signal pathways.Methods: Effects of LPS on the expression of TLR4 were tested using real-time PCR, flow cytometry and fluorescence immunostaining. Effects of LPS-induced expression/secretion of CXCL1/CXCL2 were studied using real-time PCR in cell lysates and ELISA in conditioned media of cultured uveal melanocytes. Activated NF-κB and phosphorylated MAPK signals were tested in cells with and without LPS treatment using flow cytometry. Effects of various signal inhibitors on p38, ERK1/2, JNK1/2 and NF-κB on the secretion of CXCL1/CXCL2 were tested by ELISA. The effects of neutralized antibodies of CXCL1/CXCL2 on the severity of LPS-induced uveitis were tested in a mouse model.Results: LPS stimulation increased the expression of TLR4 mRNA and protein in culture uveal melanocytes. Constitutive secretion of CXCL1/CXCL2 was detected in uveal melanocytes and was significantly increased dose- and time-dependently by LPS stimulation. LPS mainly increased the activated NF-κB and phosphorylated JNK1/2. LPS-induced expression of CXCL1/CXCL2 was blocked by NF-κB and JNK1/2 inhibitors. The severity of LPS-induced uveitis was significantly inhibited by neutralizing antibody to CXCL1/CXCL2Conclusions: This is the first report on the LPS-induced expression of CXCL1 and CXCL2 by uveal melanocytes via the activation of TLR4. These results suggest that uveal melanocytes may play a role in the immune reaction that eliminates the invading pathogens. Conversely, an excessive LPS-induced inflammatory reaction may also lead to the development of inflammatory ocular disorders, such as non-infectious uveitis.
Collapse
Affiliation(s)
- Dan-Ning Hu
- Tissue Culture Center, New York Eye and Ear Infirmary of Mount Sinai, New York, USA
- Departments of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Ruihua Zhang
- Departments of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Shen Yao
- Departments of Medicine, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Codrin E Iacob
- Departments of Pathology, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Pathology, New York Eye and Ear Infirmary of Mount Sinai, New York, USA
| | - Wei-En Yang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Richard Rosen
- Departments of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Ophthalmology, New York Eye and Ear Infirmay of Mount Sinai, New York, USA
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
9
|
Brelstaff JH, Mason M, Katsinelos T, McEwan WA, Ghetti B, Tolkovsky AM, Spillantini MG. Microglia become hypofunctional and release metalloproteases and tau seeds when phagocytosing live neurons with P301S tau aggregates. SCIENCE ADVANCES 2021; 7:eabg4980. [PMID: 34669475 PMCID: PMC8528424 DOI: 10.1126/sciadv.abg4980] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 08/26/2021] [Indexed: 05/03/2023]
Abstract
The microtubule-associated protein tau aggregates in multiple neurodegenerative diseases, causing inflammation and changing the inflammatory signature of microglia by unknown mechanisms. We have shown that microglia phagocytose live neurons containing tau aggregates cultured from P301S tau mice due to neuronal tau aggregate-induced exposure of the “eat me” signal phosphatidylserine. Here, we show that after phagocytosing tau aggregate-bearing neurons, microglia become hypophagocytic while releasing seed-competent insoluble tau aggregates. These microglia express a senescence-like phenotype, demonstrated by acidic β-galactosidase activity, secretion of paracrine senescence-associated cytokines, and maturation of matrix remodeling enzymes, results that are corroborated in P301S mouse brains and ex vivo brain slices. In particular, the nuclear factor κB–dependent activation of matrix metalloprotease 3 (MMP3/stromelysin1) was replicated in brains from patients with tauopathy. These data show that microglia that have been activated to ingest live tau aggregates-bearing neurons behave hormetically, becoming hypofunctional while acting as vectors of tau aggregate spreading.
Collapse
Affiliation(s)
- Jack H. Brelstaff
- Department of Clinical Neurosciences, Clifford Allbutt Building, University of Cambridge, Cambridge CB2 0AH, UK
| | - Matthew Mason
- Department of Clinical Neurosciences, Clifford Allbutt Building, University of Cambridge, Cambridge CB2 0AH, UK
| | - Taxiarchis Katsinelos
- Department of Clinical Neurosciences, Clifford Allbutt Building, University of Cambridge, Cambridge CB2 0AH, UK
- UK Dementia Research Institute Cambridge, Island Research Building, University of Cambridge, Cambridge CB2 0AH, UK
| | - William A. McEwan
- Department of Clinical Neurosciences, Clifford Allbutt Building, University of Cambridge, Cambridge CB2 0AH, UK
- UK Dementia Research Institute Cambridge, Island Research Building, University of Cambridge, Cambridge CB2 0AH, UK
| | - Bernardino Ghetti
- Department of Pathology and Laboratory Medicine, Indiana University, Indianapolis, IN, USA
| | - Aviva M. Tolkovsky
- Department of Clinical Neurosciences, Clifford Allbutt Building, University of Cambridge, Cambridge CB2 0AH, UK
| | - Maria Grazia Spillantini
- Department of Clinical Neurosciences, Clifford Allbutt Building, University of Cambridge, Cambridge CB2 0AH, UK
| |
Collapse
|
10
|
Perrelli A, Retta SF. Polymorphisms in genes related to oxidative stress and inflammation: Emerging links with the pathogenesis and severity of Cerebral Cavernous Malformation disease. Free Radic Biol Med 2021; 172:403-417. [PMID: 34175437 DOI: 10.1016/j.freeradbiomed.2021.06.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 06/03/2021] [Accepted: 06/22/2021] [Indexed: 02/07/2023]
Abstract
Cerebral Cavernous Malformation (CCM) is a cerebrovascular disease of genetic origin affecting 0.5% of the population and characterized by abnormally enlarged and leaky capillaries that predispose to seizures, neurological deficits, and intracerebral hemorrhage (ICH). CCM occurs sporadically or is inherited as dominant condition with incomplete penetrance and highly variable expressivity. Three disease genes have been identified: KRIT1 (CCM1), CCM2 and CCM3. Previous results demonstrated that loss-of-function mutations of CCM genes cause pleiotropic effects, including defective autophagy, altered reactive oxygen species (ROS) homeostasis, and enhanced sensitivity to oxidative stress and inflammatory events, suggesting a novel unifying pathogenetic mechanism, and raising the possibility that CCM disease onset and severity are influenced by the presence of susceptibility and modifier genes. Consistently, genome-wide association studies (GWAS) in large and homogeneous cohorts of patients sharing the familial form of CCM disease and identical mutations in CCM genes have led to the discovery of distinct genetic modifiers of major disease severity phenotypes, such as development of numerous and large CCM lesions, and susceptibility to ICH. This review deals with the identification of genetic modifiers with a significant impact on inter-individual variability in CCM disease onset and severity, including highly polymorphic genes involved in oxidative stress, inflammatory and immune responses, such as cytochrome P450 monooxygenases (CYP), matrix metalloproteinases (MMP), and Toll-like receptors (TLR), pointing to their emerging prognostic value, and opening up new perspectives for risk stratification and personalized medicine strategies.
Collapse
Affiliation(s)
- Andrea Perrelli
- Department of Clinical and Biological Sciences, University of Torino, 10043 Orbassano, Torino, Italy; CCM Italia Research Network, National Coordination Center at the Department of Clinical and Biological Sciences, University of Torino, 10043 Orbassano, Torino, Italy.
| | - Saverio Francesco Retta
- Department of Clinical and Biological Sciences, University of Torino, 10043 Orbassano, Torino, Italy; CCM Italia Research Network, National Coordination Center at the Department of Clinical and Biological Sciences, University of Torino, 10043 Orbassano, Torino, Italy.
| |
Collapse
|
11
|
MMP2 Modulates Inflammatory Response during Axonal Regeneration in the Murine Visual System. Cells 2021; 10:cells10071672. [PMID: 34359839 PMCID: PMC8307586 DOI: 10.3390/cells10071672] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/21/2021] [Accepted: 06/26/2021] [Indexed: 12/17/2022] Open
Abstract
Neuroinflammation has been put forward as a mechanism triggering axonal regrowth in the mammalian central nervous system (CNS), yet little is known about the underlying cellular and molecular players connecting these two processes. In this study, we provide evidence that MMP2 is an essential factor linking inflammation to axonal regeneration by using an in vivo mouse model of inflammation-induced axonal regeneration in the optic nerve. We show that infiltrating myeloid cells abundantly express MMP2 and that MMP2 deficiency results in reduced long-distance axonal regeneration. However, this phenotype can be rescued by restoring MMP2 expression in myeloid cells via a heterologous bone marrow transplantation. Furthermore, while MMP2 deficiency does not affect the number of infiltrating myeloid cells, it does determine the coordinated expression of pro- and anti-inflammatory molecules. Altogether, in addition to its role in axonal regeneration via resolution of the glial scar, here, we reveal a new mechanism via which MMP2 facilitates axonal regeneration, namely orchestrating the expression of pro- and anti-inflammatory molecules by infiltrating innate immune cells.
Collapse
|
12
|
Ocular TGF- β, Matrix Metalloproteinases, and TIMP-1 Increase with the Development and Progression of Diabetic Retinopathy in Type 2 Diabetes Mellitus. Mediators Inflamm 2021; 2021:9811361. [PMID: 34257518 PMCID: PMC8257377 DOI: 10.1155/2021/9811361] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 06/04/2021] [Indexed: 01/20/2023] Open
Abstract
Diabetic retinopathy (DR) is a sight-threatening late complication of diabetes mellitus (DM). Even though its pathophysiology has not been fully elucidated, several studies suggested a role for transforming growth factor- (TGF-) β, matrix metalloproteinases (MMPs), and tissue inhibitors of matrix metalloproteinase (TIMP) in the onset and progression of the disease. Consequently, the aim of this study was to analyze the concentrations of TGF-β1, TGF-β2, TGF-β3, MMP-3, MMP-9, and TIMP-1 in patients with different stages of DR in order to identify stage-specific changes in their concentrations during the progression of the disease. Serum and aqueous humor (AH) samples were collected during intraocular surgery, and eyes were classified into the following groups: healthy controls (n = 17), diabetic patients with non-apparent DR (n = 23), mild/moderate nonproliferative DR (NPDR) (n = 13), and advanced NPDR/proliferative DR (PDR) without vitreal hemorrhage (n = 14). None of the patients had been under anti-VEGF or laser treatment within six months prior to surgery. In the AH, TGF-β1 levels increased in advanced NPDR/PDR by a factor of 5.5 compared to the control group. Similarly, an increase in MMP-3 and TIMP-1 levels in the AH was evident in the later stages of DR, corresponding to a 7.7- and 2.4-fold increase compared to the control group, respectively, whereas serum levels of the studied proteins remained similar. In conclusion, increased concentrations of TGF-β1, MMP-3, and TIMP-1 in the AH, but not in the serum, in advanced NPDR/PDR indicate that the intraocular regulation for these cytokines is independent of the systemic one and suggest their involvement in the progression of DR.
Collapse
|
13
|
Craenen K, Verslegers M, Callaerts-Vegh Z, Craeghs L, Buset J, Govaerts K, Neefs M, Gsell W, Baatout S, D'Hooge R, Himmelreich U, Moons L, Benotmane MA. Folic Acid Fortification Prevents Morphological and Behavioral Consequences of X-Ray Exposure During Neurulation. Front Behav Neurosci 2021; 14:609660. [PMID: 33488367 PMCID: PMC7820780 DOI: 10.3389/fnbeh.2020.609660] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 12/08/2020] [Indexed: 12/02/2022] Open
Abstract
Previous studies suggested a causal link between pre-natal exposure to ionizing radiation and birth defects such as microphthalmos and exencephaly. In mice, these defects arise primarily after high-dose X-irradiation during early neurulation. However, the impact of sublethal (low) X-ray doses during this early developmental time window on adult behavior and morphology of central nervous system structures is not known. In addition, the efficacy of folic acid (FA) in preventing radiation-induced birth defects and persistent radiation-induced anomalies has remained unexplored. To assess the efficacy of FA in preventing radiation-induced defects, pregnant C57BL6/J mice were X-irradiated at embryonic day (E)7.5 and were fed FA-fortified food. FA partially prevented radiation-induced (1.0 Gy) anophthalmos, exencephaly and gastroschisis at E18, and reduced the number of pre-natal deaths, fetal weight loss and defects in the cervical vertebrae resulting from irradiation. Furthermore, FA food fortification counteracted radiation-induced impairments in vision and olfaction, which were evidenced after exposure to doses ≥0.1 Gy. These findings coincided with the observation of a reduction in thickness of the retinal ganglion cell and nerve fiber layer, and a decreased axial length of the eye following exposure to 0.5 Gy. Finally, MRI studies revealed a volumetric decrease of the hippocampus, striatum, thalamus, midbrain and pons following 0.5 Gy irradiation, which could be partially ameliorated after FA food fortification. Altogether, our study is the first to offer detailed insights into the long-term consequences of X-ray exposure during neurulation, and supports the use of FA as a radioprotectant and antiteratogen to counter the detrimental effects of X-ray exposure during this crucial period of gestation.
Collapse
Affiliation(s)
- Kai Craenen
- Radiobiology Unit, Interdisciplinary Biosciences, Institute for Environment, Health and Safety, Belgian Nuclear Research Centre (Studiecentrum voor Kernenergie; Centre d'étude de l'énergie nucléaire), Mol, Belgium
- Laboratory of Neural Circuit Development and Regeneration, Animal Physiology and Neurobiology Section, Department of Biology, Faculty of Science, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Mieke Verslegers
- Radiobiology Unit, Interdisciplinary Biosciences, Institute for Environment, Health and Safety, Belgian Nuclear Research Centre (Studiecentrum voor Kernenergie; Centre d'étude de l'énergie nucléaire), Mol, Belgium
| | - Zsuzsanna Callaerts-Vegh
- Laboratory of Biological Psychology, Faculty of Psychology and Educational Sciences, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Livine Craeghs
- Radiobiology Unit, Interdisciplinary Biosciences, Institute for Environment, Health and Safety, Belgian Nuclear Research Centre (Studiecentrum voor Kernenergie; Centre d'étude de l'énergie nucléaire), Mol, Belgium
- Laboratory of Neural Circuit Development and Regeneration, Animal Physiology and Neurobiology Section, Department of Biology, Faculty of Science, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Jasmine Buset
- Radiobiology Unit, Interdisciplinary Biosciences, Institute for Environment, Health and Safety, Belgian Nuclear Research Centre (Studiecentrum voor Kernenergie; Centre d'étude de l'énergie nucléaire), Mol, Belgium
| | - Kristof Govaerts
- Molecular Small Animal Imaging Center, Biomedical MRI Unit, Department of Imaging and Pathology, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Mieke Neefs
- Radiobiology Unit, Interdisciplinary Biosciences, Institute for Environment, Health and Safety, Belgian Nuclear Research Centre (Studiecentrum voor Kernenergie; Centre d'étude de l'énergie nucléaire), Mol, Belgium
| | - Willy Gsell
- Molecular Small Animal Imaging Center, Biomedical MRI Unit, Department of Imaging and Pathology, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Sarah Baatout
- Radiobiology Unit, Interdisciplinary Biosciences, Institute for Environment, Health and Safety, Belgian Nuclear Research Centre (Studiecentrum voor Kernenergie; Centre d'étude de l'énergie nucléaire), Mol, Belgium
| | - Rudi D'Hooge
- Laboratory of Biological Psychology, Faculty of Psychology and Educational Sciences, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Uwe Himmelreich
- Molecular Small Animal Imaging Center, Biomedical MRI Unit, Department of Imaging and Pathology, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Lieve Moons
- Radiobiology Unit, Interdisciplinary Biosciences, Institute for Environment, Health and Safety, Belgian Nuclear Research Centre (Studiecentrum voor Kernenergie; Centre d'étude de l'énergie nucléaire), Mol, Belgium
- Laboratory of Neural Circuit Development and Regeneration, Animal Physiology and Neurobiology Section, Department of Biology, Faculty of Science, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Mohammed Abderrafi Benotmane
- Radiobiology Unit, Interdisciplinary Biosciences, Institute for Environment, Health and Safety, Belgian Nuclear Research Centre (Studiecentrum voor Kernenergie; Centre d'étude de l'énergie nucléaire), Mol, Belgium
| |
Collapse
|
14
|
García-Onrubia L, Valentín-Bravo FJ, Coco-Martin RM, González-Sarmiento R, Pastor JC, Usategui-Martín R, Pastor-Idoate S. Matrix Metalloproteinases in Age-Related Macular Degeneration (AMD). Int J Mol Sci 2020; 21:ijms21165934. [PMID: 32824762 PMCID: PMC7460693 DOI: 10.3390/ijms21165934] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/11/2020] [Accepted: 08/12/2020] [Indexed: 12/13/2022] Open
Abstract
Age-related macular degeneration (AMD) is a complex, multifactorial and progressive retinal disease affecting millions of people worldwide. In developed countries, it is the leading cause of vision loss and legal blindness among the elderly. Although the pathogenesis of AMD is still barely understood, recent studies have reported that disorders in the regulation of the extracellular matrix (ECM) play an important role in its etiopathogenesis. The dynamic metabolism of the ECM is closely regulated by matrix metalloproteinases (MMPs) and the tissue inhibitors of metalloproteinases (TIMPs). The present review focuses on the crucial processes that occur at the level of the Bruch’s membrane, with special emphasis on MMPs, TIMPs, and the polymorphisms associated with increased susceptibility to AMD development. A systematic literature search was performed, covering the years 1990–2020, using the following keywords: AMD, extracellular matrix, Bruch’s membrane, MMPs, TIMPs, and MMPs polymorphisms in AMD. In both early and advanced AMD, the pathological dynamic changes of ECM structural components are caused by the dysfunction of specific regulators and by the influence of other regulatory systems connected with both genetic and environmental factors. Better insight into the pathological role of MMP/TIMP complexes may lead to the development of new strategies for AMD treatment and prevention.
Collapse
Affiliation(s)
- Luis García-Onrubia
- Clinical University Hospital of Valladolid, Av. Ramón y Cajal, 3, 47003 Valladolid, Spain; (L.G.-O.); (F.J.V.-B.); (J.C.P.)
| | - Fco. Javier Valentín-Bravo
- Clinical University Hospital of Valladolid, Av. Ramón y Cajal, 3, 47003 Valladolid, Spain; (L.G.-O.); (F.J.V.-B.); (J.C.P.)
| | - Rosa M. Coco-Martin
- Institute of Applied Ophthalmobiology (IOBA), University of Valladolid, 47011 Valladolid, Spain;
- Cooperative Health Network for Research in Ophthalmology (Oftared), National Institute of Health Carlos III, ISCIII, 28040 Madrid, Spain
| | - Rogelio González-Sarmiento
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain;
- Institute of Molecular and Cellular Biology of Cancer (IBMCC), University of Salamanca—CSIC, 37007 Salamanca, Spain
| | - J. Carlos Pastor
- Clinical University Hospital of Valladolid, Av. Ramón y Cajal, 3, 47003 Valladolid, Spain; (L.G.-O.); (F.J.V.-B.); (J.C.P.)
- Institute of Applied Ophthalmobiology (IOBA), University of Valladolid, 47011 Valladolid, Spain;
- Cooperative Health Network for Research in Ophthalmology (Oftared), National Institute of Health Carlos III, ISCIII, 28040 Madrid, Spain
| | - Ricardo Usategui-Martín
- Institute of Applied Ophthalmobiology (IOBA), University of Valladolid, 47011 Valladolid, Spain;
- Correspondence: (R.U.-M.); (S.P.-I.)
| | - Salvador Pastor-Idoate
- Clinical University Hospital of Valladolid, Av. Ramón y Cajal, 3, 47003 Valladolid, Spain; (L.G.-O.); (F.J.V.-B.); (J.C.P.)
- Institute of Applied Ophthalmobiology (IOBA), University of Valladolid, 47011 Valladolid, Spain;
- Cooperative Health Network for Research in Ophthalmology (Oftared), National Institute of Health Carlos III, ISCIII, 28040 Madrid, Spain
- Correspondence: (R.U.-M.); (S.P.-I.)
| |
Collapse
|
15
|
Lefevere E, Salinas‐Navarro M, Andries L, Noterdaeme L, Etienne I, Van Wonterghem E, Vinckier S, Davis BM, Van Bergen T, Van Hove I, Movahedi K, Vandenbroucke RE, Moons L, De Groef L. Tightening the retinal glia limitans attenuates neuroinflammation after optic nerve injury. Glia 2020; 68:2643-2660. [DOI: 10.1002/glia.23875] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 05/20/2020] [Accepted: 06/04/2020] [Indexed: 12/14/2022]
Affiliation(s)
- Evy Lefevere
- Neural Circuit Development and Regeneration Research Group, Department of Biology KU Leuven Leuven Belgium
- Leuven Brain Institute (LBI) KU Leuven Leuven Belgium
| | - Manuel Salinas‐Navarro
- Neural Circuit Development and Regeneration Research Group, Department of Biology KU Leuven Leuven Belgium
- Leuven Brain Institute (LBI) KU Leuven Leuven Belgium
| | - Lien Andries
- Neural Circuit Development and Regeneration Research Group, Department of Biology KU Leuven Leuven Belgium
- Leuven Brain Institute (LBI) KU Leuven Leuven Belgium
| | - Lut Noterdaeme
- Neural Circuit Development and Regeneration Research Group, Department of Biology KU Leuven Leuven Belgium
| | | | - Elien Van Wonterghem
- Barriers in Inflammation Lab VIB Center for Inflammation Research Ghent Belgium
- Department of Biomedical Molecular Biology Ghent University Ghent Belgium
| | - Stefan Vinckier
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, and Department of Oncology and Leuven Cancer Institute (LKI) VIB and KU Leuven Leuven Belgium
| | - Benjamin M. Davis
- Glaucoma and Retinal Neurodegeneration Research, Visual Neuroscience UCL Institute of Ophthalmology London UK
| | | | - Inge Van Hove
- Neural Circuit Development and Regeneration Research Group, Department of Biology KU Leuven Leuven Belgium
- Oxurion NV Leuven Belgium
| | - Kiavash Movahedi
- Myeloid Cell Immunology Lab VIB Center for Inflammation Research Brussels Belgium
- Lab of Cellular and Molecular Immunology Vrije Universiteit Brussel Brussels Belgium
| | - Roosmarijn E. Vandenbroucke
- Barriers in Inflammation Lab VIB Center for Inflammation Research Ghent Belgium
- Department of Biomedical Molecular Biology Ghent University Ghent Belgium
| | - Lieve Moons
- Neural Circuit Development and Regeneration Research Group, Department of Biology KU Leuven Leuven Belgium
- Leuven Brain Institute (LBI) KU Leuven Leuven Belgium
| | - Lies De Groef
- Neural Circuit Development and Regeneration Research Group, Department of Biology KU Leuven Leuven Belgium
- Leuven Brain Institute (LBI) KU Leuven Leuven Belgium
| |
Collapse
|
16
|
Zhou J, Yang J, Dai M, Lin D, Zhang R, Liu H, Yu A, Vakal S, Wang Y, Li X. A combination of inhibiting microglia activity and remodeling gut microenvironment suppresses the development and progression of experimental autoimmune uveitis. Biochem Pharmacol 2020; 180:114108. [PMID: 32569628 DOI: 10.1016/j.bcp.2020.114108] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 06/17/2020] [Accepted: 06/17/2020] [Indexed: 12/11/2022]
Abstract
Noninfectious (autoimmune and immune-mediated) uveitis is an ocular inflammatory disease which can lead to blindness in severe cases. Due to the potential side effects of first-line drugs for clinical uveitis, novel drugs and targets against uveitis are still urgently needed. In the present study, using rat experimental autoimmune uveitis (EAU) model, we first found that minocycline treatment can substantially inhibit the development of EAU and improve the retinal function by suppressing the retinal microglial activation, and block the infiltration of inflammatory cells, including Th17, into the retina by decreasing the major histocompatibility complex class II (MHC II) expression in resident and infiltrating cells. Moreover, we demonstrated that minocycline treatment can remodel the gut microenvironment of EAU rats by restoring the relative abundance of Ruminococcus bromii, Streptococcus hyointestinalis, and Desulfovibrio sp. ABHU2SB and promoting a functional shift in the gut via reversing the levels of L-proline, allicin, aceturic acid, xanthine, and leukotriene B4, and especially increasing the production of propionic acid, histamine, and pantothenic acid. At last, we revealed that minocycline treatment can significantly attenuate the progression of EAU after inflammation onset, which may be explained by the role of minocycline in the remodeling of the gut microenvironment since selective elimination of retinal microglia on the later stages of EAU was shown to have little effect. These data clearly demonstrated that inhibition of microglial activation and remodeling of the gut microenvironment can suppress the development and progression of experimental autoimmune uveitis. Considering the excellent safety profile of minocycline in multiple clinical experiments, we suggest that minocycline may have therapeutic implications for clinical uveitis.
Collapse
Affiliation(s)
- Jianhong Zhou
- School of Ophthalmology & Optometry and Eye Hospital, Institute of Biomedical Engineering, Wenzhou Medical University, Wenzhou 325027, Zhejiang, China; State Key Laboratory of Optometry & Vision Science, Wenzhou 325027, Zhejiang, China
| | - Jingjing Yang
- School of Ophthalmology & Optometry and Eye Hospital, Institute of Biomedical Engineering, Wenzhou Medical University, Wenzhou 325027, Zhejiang, China; State Key Laboratory of Optometry & Vision Science, Wenzhou 325027, Zhejiang, China
| | - Mali Dai
- School of Ophthalmology & Optometry and Eye Hospital, Institute of Biomedical Engineering, Wenzhou Medical University, Wenzhou 325027, Zhejiang, China; State Key Laboratory of Optometry & Vision Science, Wenzhou 325027, Zhejiang, China
| | - Dan Lin
- School of Ophthalmology & Optometry and Eye Hospital, Institute of Biomedical Engineering, Wenzhou Medical University, Wenzhou 325027, Zhejiang, China; State Key Laboratory of Optometry & Vision Science, Wenzhou 325027, Zhejiang, China
| | - Renshu Zhang
- School of Ophthalmology & Optometry and Eye Hospital, Institute of Biomedical Engineering, Wenzhou Medical University, Wenzhou 325027, Zhejiang, China; State Key Laboratory of Optometry & Vision Science, Wenzhou 325027, Zhejiang, China
| | - Hui Liu
- School of Ophthalmology & Optometry and Eye Hospital, Institute of Biomedical Engineering, Wenzhou Medical University, Wenzhou 325027, Zhejiang, China; State Key Laboratory of Optometry & Vision Science, Wenzhou 325027, Zhejiang, China
| | - Ailing Yu
- School of Ophthalmology & Optometry and Eye Hospital, Institute of Biomedical Engineering, Wenzhou Medical University, Wenzhou 325027, Zhejiang, China; State Key Laboratory of Optometry & Vision Science, Wenzhou 325027, Zhejiang, China
| | - Serhii Vakal
- Structural Bioinformatics Laboratory, Biochemistry, Åbo Akademi University, Turku 20541, Finland
| | - Yuqin Wang
- School of Ophthalmology & Optometry and Eye Hospital, Institute of Biomedical Engineering, Wenzhou Medical University, Wenzhou 325027, Zhejiang, China; State Key Laboratory of Optometry & Vision Science, Wenzhou 325027, Zhejiang, China.
| | - Xingyi Li
- School of Ophthalmology & Optometry and Eye Hospital, Institute of Biomedical Engineering, Wenzhou Medical University, Wenzhou 325027, Zhejiang, China; State Key Laboratory of Optometry & Vision Science, Wenzhou 325027, Zhejiang, China.
| |
Collapse
|
17
|
Chaudhary R, Scott RAH, Wallace G, Berry M, Logan A, Blanch RJ. Inflammatory and Fibrogenic Factors in Proliferative Vitreoretinopathy Development. Transl Vis Sci Technol 2020; 9:23. [PMID: 32742753 PMCID: PMC7357815 DOI: 10.1167/tvst.9.3.23] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Purpose Proliferative vitreoretinopathy (PVR) occurs in 5%-10% of rhegmatogenous retinal detachment cases and is the principle cause for failure of retinal reattachment surgery. Although there are a number of surgical adjunctive agents available for preventing the development of PVR, all have limited efficacy. Discovering predictive molecular biomarkers to determine the probability of PVR development after retinal reattachment surgery will allow better patient stratification for more targeted drug evaluations. Methods Narrative literature review. Results We provide a summary of the inflammatory and fibrogenic factors found in ocular fluid samples during the development of retinal detachment and PVR and discuss their possible use as molecular PVR predictive biomarkers. Conclusions Studies monitoring the levels of the above factors have found that few if any have predictive biomarker value, suggesting that widening the phenotype of potential factors and a combinatorial approach are required to determine predictive biomarkers for PVR. Translational Relevance The identification of relevant biomarkers relies on an understanding of disease signaling pathways derived from basic science research. We discuss the extent to which those molecules identified as biomarkers and predictors of PVR relate to disease pathogenesis and could function as useful disease predictors. (http://www.umin.ac.jp/ctr/ number, UMIN000005604).
Collapse
Affiliation(s)
- Rishika Chaudhary
- Academic Unit of Ophthalmology, Birmingham and Midland Eye Centre, Birmingham, UK.,Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK.,NIHR Surgical Reconstruction and Microbiology Centre, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | | | - Graham Wallace
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Martin Berry
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Ann Logan
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK.,NIHR Surgical Reconstruction and Microbiology Centre, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Richard J Blanch
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK.,NIHR Surgical Reconstruction and Microbiology Centre, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK.,Department of Ophthalmology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK.,Academic Unit of Military Surgery and Trauma, Royal Centre for Defence Medicine, Birmingham, UK
| |
Collapse
|
18
|
Zeng W, Zhang Y, Duan F, Lin T, Liu X, Li D, Wu K. Lipopolysaccharide enhances human herpesvirus 1 replication and IL-6 release in epithelial cells. Microb Pathog 2020; 140:103961. [PMID: 31904451 DOI: 10.1016/j.micpath.2019.103961] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 12/02/2019] [Accepted: 12/31/2019] [Indexed: 12/24/2022]
Abstract
OBJECTIVE To investigate the effect of lipopolysaccharide (LPS) on human herpesvirus 1 (HHV-1) infection in epithelial cells. METHODS Two strains of HHV-1, HHV-1 F strain (HHV-1f) and HHV-1 strain-H129 with GFP knock-in (HHV-g4), were used to infect HCE-T and VERO cells at MOIs of 0.04 and 0.02, respectively. After 1 h, 0, 10, 50, and 100 μg/ml LPS was added to serum-free medium and the cells were cultured for up to 24 h. GFP fluorescence of HHV-g4 in cells was examined under a fluorescence microscope and imaged. HHV-1f titer was determined by quantitative real-time polymerase chain reaction (qPCR) in HCE-T cells and plaque assays in VERO cells. The expression of the viral ICP4 protein of HHV-1f was detected by Western blot assay. IL-6 and IL-10 levels in culture medium were determined by enzyme-linked immunosorbent assay (ELISA). RESULTS Similar changes but at different degrees were found in HCE-T and VERO cells that were infected with HHV-1. GFP fluorescence of HHV-g4 and cell lesions increased in a dose-dependent manner. Virus titer was also enhanced by LPS stimulation in HCE-T and VERO cells. ICP4 expression was promoted at higher LPS concentrations (P = 0.04). In addition, viral infection resulted in increased expression of IL-6 in a dose-dependent manner at 12 and 24 h (P = 0.01), while IL-10 expression was unaffected by either HHV-1 infection or LPS stimulation. CONCLUSION LPS promotes HHV-1 infection in epithelial cells, which suggests that gram-negative bacteria on ocular surfaces may aggravate HHV-1 infection.
Collapse
Affiliation(s)
- Weiting Zeng
- Zhongshan Ophthalmic Center, State Key Laboratory of Ophthalmology, the Key Lab of Ophthalmology and Visual Science of Guangdong, Sun Yat-sen University, Guangzhou, China
| | - Yafang Zhang
- Department of Ophthalmology, Hubei University of Science and Technology, Xianning, China
| | - Fang Duan
- Zhongshan Ophthalmic Center, State Key Laboratory of Ophthalmology, the Key Lab of Ophthalmology and Visual Science of Guangdong, Sun Yat-sen University, Guangzhou, China
| | - Tianlan Lin
- Zhongshan Ophthalmic Center, State Key Laboratory of Ophthalmology, the Key Lab of Ophthalmology and Visual Science of Guangdong, Sun Yat-sen University, Guangzhou, China
| | - Xiuping Liu
- Zhongshan Ophthalmic Center, State Key Laboratory of Ophthalmology, the Key Lab of Ophthalmology and Visual Science of Guangdong, Sun Yat-sen University, Guangzhou, China
| | - Dai Li
- Department of Ophthalmology, Hubei University of Science and Technology, Xianning, China.
| | - Kaili Wu
- Zhongshan Ophthalmic Center, State Key Laboratory of Ophthalmology, the Key Lab of Ophthalmology and Visual Science of Guangdong, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
19
|
Sergeys J, Etienne I, Van Hove I, Lefevere E, Stalmans I, Feyen JHM, Moons L, Van Bergen T. Longitudinal In Vivo Characterization of the Streptozotocin-Induced Diabetic Mouse Model: Focus on Early Inner Retinal Responses. Invest Ophthalmol Vis Sci 2019; 60:807-822. [PMID: 30811545 DOI: 10.1167/iovs.18-25372] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose The goal of this study was to perform an extensive temporal characterization of the early pathologic processes in the streptozotocin (STZ)-induced diabetic retinopathy (DR) mouse model, beyond the vascular phenotype, and to investigate the potential of clinically relevant compounds in attenuating these processes. Methods Visual acuity and contrast sensitivity (CS) were studied in the mouse STZ model until 24 weeks postdiabetes onset. ERG, spectral domain optical coherence tomography (SD-OCT), leukostasis, and immunohistochemistry were applied to investigate neurodegeneration, inflammation, and gliosis during early-, mid- and late-phase diabetes. Aflibercept or triamcinolone acetonide (TAAC) was administered to investigate their efficacy on the aforementioned processes. Results Visual acuity and CS loss started at 4 and 18 weeks postdiabetes onset, respectively, and progressively declined over time. ERG amplitudes were diminished and OP latencies increased after 6 weeks, whereas SD-OCT revealed retinal thinning from 4 weeks postdiabetes. Immunohistochemical analyses linked these findings to retinal ganglion and cholinergic amacrine cell loss at 4 and 8 weeks postdiabetes onset, respectively, which was further decreased after aflibercept administration. The number of adherent leukocytes was augmented after 2 weeks, whereas increased micro- and macroglia reactivity was present from 4 weeks postdiabetes. Aflibercept or TAAC showed improved efficacy on inflammation and gliosis. Conclusions STZ-induced diabetic mice developed early pathologic DR hallmarks, from which inflammation seemed the initial trigger, leading to further development of functional and morphologic retinal changes. These findings indicate that the mouse STZ model is suitable to study novel integrative non-vascular therapies to treat early DR.
Collapse
Affiliation(s)
- Jurgen Sergeys
- Neural Circuit Development and Regeneration Research Group, Department of Biology, Zoological Institute, KU Leuven, Leuven, Belgium
| | | | - Inge Van Hove
- Neural Circuit Development and Regeneration Research Group, Department of Biology, Zoological Institute, KU Leuven, Leuven, Belgium.,Oxurion NV, Leuven, Belgium
| | - Evy Lefevere
- Neural Circuit Development and Regeneration Research Group, Department of Biology, Zoological Institute, KU Leuven, Leuven, Belgium
| | - Ingeborg Stalmans
- Laboratory of Experimental Ophthalmology, Department of Neurosciences, O&N II, KU Leuven, Leuven, Belgium
| | | | - Lieve Moons
- Neural Circuit Development and Regeneration Research Group, Department of Biology, Zoological Institute, KU Leuven, Leuven, Belgium
| | | |
Collapse
|
20
|
Critical Role of Monocyte Recruitment in Optic Nerve Damage Induced by Experimental Optic Neuritis. Mol Neurobiol 2019; 56:7458-7472. [PMID: 31044366 DOI: 10.1007/s12035-019-1608-0] [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: 11/21/2018] [Accepted: 04/12/2019] [Indexed: 12/13/2022]
Abstract
Neuroinflammatory diseases are characterized by blood-brain barrier disruption (BBB) and leukocyte infiltration. We investigated the involvement of monocyte recruitment in visual pathway damage provoked by primary optic neuritis (ON) induced by a microinjection of bacterial lipopolysaccharide (LPS) into the optic nerve from male Wistar rats. Increased Evans blue extravasation and cellularity were observed at 6 h post-LPS injection. In WT-GFPþ/WT chimeric rat optic nerves, the presence of GFP(+) neutrophils and GFP(+) monocytes, and in wild-type rat optic nerves, an increase in CD11b+CD45low and CD11b+CD45high cell number, were observed at 24 h post-LPS. Gamma-irradiation did not affect the increase in BBB permeability, but significantly lessened the decrease in pupil light reflex (PLR), and retinal ganglion cell (RGC) number induced by LPS. At 6 h post-LPS, an increase in chemokine (C-C motif) ligand 2 (CCL2) immunoreactivity co-localized with neutrophils (but not microglia/macrophages or astrocytes) was observed, while at 24 h post-injection, an increase in Iba-1-immunoreactivity and its co-localization with CCL2 became evident. The co-injection of LPS with bindarit (a CCL2 synthesis inhibitor) lessened the effect of LPS on PLR, and RGC loss. The treatment with etoposide or gadolinium chloride that significantly decreased peripheral monocyte (but not neutrophil or lymphocyte) percentage decreased the effect of LPS on PLR, and RGC number. Moreover, a negative correlation between PRL and monocyte (but not lymphocyte or neutrophil) percentage was observed at 7 days post-LPS. Taken together, these results support that monocytes are key players in the initial events that take place during primary ON.
Collapse
|
21
|
Caolo V, Roblain Q, Lecomte J, Carai P, Peters L, Cuijpers I, Robinson EL, Derks K, Sergeys J, Noël A, Jones EAV, Moons L, Heymans S. Resistance to retinopathy development in obese, diabetic and hypertensive ZSF1 rats: an exciting model to identify protective genes. Sci Rep 2018; 8:11922. [PMID: 30093686 PMCID: PMC6085379 DOI: 10.1038/s41598-018-29812-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 07/17/2018] [Indexed: 12/11/2022] Open
Abstract
Diabetic retinopathy (DR) is one of the major complications of diabetes, which eventually leads to blindness. Up to date, no animal model has yet shown all the co-morbidities often observed in DR patients. Here, we investigated whether obese 42 weeks old ZSF1 rat, which spontaneously develops diabetes, hypertension and obesity, would be a suitable model to study DR. Although arteriolar tortuosity increased in retinas from obese as compared to lean (hypertensive only) ZSF1 rats, vascular density pericyte coverage, microglia number, vascular morphology and retinal thickness were not affected by diabetes. These results show that, despite high glucose levels, obese ZSF1 rats did not develop DR. Such observations prompted us to investigate whether the expression of genes, possibly able to contain DR development, was affected. Accordingly, mRNA sequencing analysis showed that genes (i.e. Npy and crystallins), known to have a protective role, were upregulated in retinas from obese ZSF1 rats. Lack of retina damage, despite obesity, hypertension and diabetes, makes the 42 weeks of age ZSF1 rats a suitable animal model to identify genes with a protective function in DR. Further characterisation of the identified genes and downstream pathways could provide more therapeutic targets for the treat DR.
Collapse
Affiliation(s)
- Vincenza Caolo
- Department of Cardiovascular Sciences, Centre for Molecular and Vascular Biology, KU Leuven, Belgium.
| | - Quentin Roblain
- Department of Cardiology, CARIM School for Cardiovascular Diseases Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands.,Laboratory of Tumor and Development Biology, GIGA-Cancer, University of Liège, Liège, Belgium
| | - Julie Lecomte
- Laboratory of Tumor and Development Biology, GIGA-Cancer, University of Liège, Liège, Belgium
| | - Paolo Carai
- Department of Cardiovascular Sciences, Centre for Molecular and Vascular Biology, KU Leuven, Belgium
| | - Linsey Peters
- Department of Cardiology, CARIM School for Cardiovascular Diseases Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
| | - Ilona Cuijpers
- Department of Cardiovascular Sciences, Centre for Molecular and Vascular Biology, KU Leuven, Belgium.,Department of Cardiology, CARIM School for Cardiovascular Diseases Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
| | - Emma Louise Robinson
- Department of Cardiology, CARIM School for Cardiovascular Diseases Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
| | - Kasper Derks
- Department of Genetics and Cell Biology, CARIM School for Cardiovascular Diseases Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
| | - Jurgen Sergeys
- Laboratory of Neural Circuit Development and Regeneration, Animal Physiology and Neurobiology Section, Department of Biology, KU Leuven, Leuven, Belgium
| | - Agnès Noël
- Laboratory of Tumor and Development Biology, GIGA-Cancer, University of Liège, Liège, Belgium
| | - Elizabeth A V Jones
- Department of Cardiovascular Sciences, Centre for Molecular and Vascular Biology, KU Leuven, Belgium
| | - Lieve Moons
- Laboratory of Neural Circuit Development and Regeneration, Animal Physiology and Neurobiology Section, Department of Biology, KU Leuven, Leuven, Belgium
| | - Stephane Heymans
- Department of Cardiovascular Sciences, Centre for Molecular and Vascular Biology, KU Leuven, Belgium.,Department of Cardiology, CARIM School for Cardiovascular Diseases Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands.,The Netherlands Heart Institute, Nl-HI, Utrecht, The Netherlands
| |
Collapse
|
22
|
Matrix metalloproteinases as regulators of inflammatory processes. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:2036-2042. [DOI: 10.1016/j.bbamcr.2017.05.010] [Citation(s) in RCA: 125] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 05/06/2017] [Accepted: 05/09/2017] [Indexed: 12/26/2022]
|
23
|
Rapid monocyte infiltration following retinal detachment is dependent on non-canonical IL6 signaling through gp130. J Neuroinflammation 2017. [PMID: 28645275 PMCID: PMC5481880 DOI: 10.1186/s12974-017-0886-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Retinal detachment (RD) can lead to proliferative vitreoretinopathy (PVR), a leading cause of intractable vision loss. PVR is associated with a cytokine storm involving common proinflammatory molecules like IL6, but little is known about the source and downstream signaling of IL6 and the consequences for the retina. Here, we investigated the early immune response and resultant cytokine signaling following RD in mice. METHODS RD was induced in C57BL/6 J and IL6 knockout mice, and the resulting inflammatory response was examined using immunohistochemistry and flow cytometry. Cytokines and signaling proteins of vitreous and retinas were quantified by multiple cytokine arrays and Western blotting. To attempt to block IL6 signaling, a neutralizing antibody of IL6 receptor α (IL6Rα) or IL6 receptor β (gp-130) was injected intravitreally immediately after RD. RESULTS Within one day of RD, bone marrow-derived Cd11b + monocytes had extravasated from the vasculature and lined the vitreal surface of the retina, while the microglia, the resident macrophages of the retina, were relatively unperturbed. Cytokine arrays and Western blot analysis revealed that this sterile inflammation did not cause activation of IL6 signaling in the neurosensory retina, but rather only in the vitreous and aqueous humor. Monocyte infiltration was inhibited by blocking gp130, but not by IL6 knockout or IL6Rα blockade. CONCLUSIONS Together, our results demonstrate that monocytes are the primary immune cell mediating the cytokine storm following RD, and that any resulting retinal damage is unlikely to be a direct result of retinal IL6 signaling, but rather gp130-mediated signaling in the monocytes themselves. These results suggest that RD should be treated immediately, and that gp130-directed therapies may prevent PVR and promote retinal healing.
Collapse
|