1
|
Chaudhary JK, Danga AK, Kumari A, Bhardwaj A, Rath PC. Role of chemokines in aging and age-related diseases. Mech Ageing Dev 2024; 223:112009. [PMID: 39631472 DOI: 10.1016/j.mad.2024.112009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 11/21/2024] [Accepted: 11/25/2024] [Indexed: 12/07/2024]
Abstract
Chemokines (chemotactic cytokines) play essential roles in developmental process, immune cell trafficking, inflammation, immunity, angiogenesis, cellular homeostasis, aging, neurodegeneration, and tumorigenesis. Chemokines also modulate response to immunotherapy, and consequently influence the therapeutic outcome. The mechanisms underlying these processes are accomplished by interaction of chemokines with their cognate cell surface G protein-coupled receptors (GPCRs) and subsequent cellular signaling pathways. Chemokines play crucial role in influencing aging process and age-related diseases across various tissues and organs, primarily through inflammatory responses (inflammaging), recruitment of macrophages, and orchestrated trafficking of other immune cells. Chemokines are categorized in four distinct groups based on the position and number of the N-terminal cysteine residues; namely, the CC, CXC, CX3C, and (X)C. They mediate inflammatory responses, and thereby considerably impact aging process across multiple organ-systems. Therefore, understanding the underlying mechanisms mediated by chemokines may be of crucial importance in delaying and/or modulating the aging process and preventing age-related diseases. In this review, we highlight recent progress accomplished towards understanding the role of chemokines and their cellular signaling pathways involved in aging and age-relaed diseases of various organs. Moreover, we explore potential therapeutic strategies involving anti-chemokines and chemokine receptor antagonists aimed at reducing aging and mitigating age-related diseases. One of the modern methods in this direction involves use of chemokine receptor antagonists and anti-chemokines, which suppress the pro-inflammatory response, thereby helping in resolution of inflammation. Considering the wide-spectrum of functional involvements of chemokines in aging and associated diseases, several clinical trials are being conducted to develop therapeutic approaches using anti-chemokine and chemokine receptor antagonists to improve life span and promote healthy aging.
Collapse
Affiliation(s)
- Jitendra Kumar Chaudhary
- Molecular Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India; Department of Zoology, Shivaji College, University of Delhi, New Delhi 110027, India.
| | - Ajay Kumar Danga
- Molecular Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India.
| | - Anita Kumari
- Molecular Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India.
| | - Akshay Bhardwaj
- Regional Centre for Biotechnology, 3rd Milestone, Faridabad-Gurugram Expressway, Faridabad Road, Faridabad, Haryana 121001, India.
| | - Pramod C Rath
- Molecular Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India.
| |
Collapse
|
2
|
Guo D, Zhu W, Qiu H. C-C Motif Chemokine Ligand 2 and Chemokine Receptor 2 in Cardiovascular and Neural Aging and Aging-Related Diseases. Int J Mol Sci 2024; 25:8794. [PMID: 39201480 PMCID: PMC11355023 DOI: 10.3390/ijms25168794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 09/02/2024] Open
Abstract
Aging is a prominent risk factor for numerous chronic diseases. Understanding the shared mechanisms of aging can aid in pinpointing therapeutic targets for age-related disorders. Chronic inflammation has emerged as a pivotal mediator of aging and a determinant in various age-related chronic conditions. Recent findings indicate that C-C motif chemokine ligand 2 and receptor 2 (CCL2-CCR2) signaling, an important physiological modulator in innate immune response and inflammatory defense, plays a crucial role in aging-related disorders and is increasingly recognized as a promising therapeutic target, highlighting its significance. This review summarizes recent advances in the investigation of CCL2-CCR2 signaling in cardiovascular and neural aging, as well as in various aging-related disorders. It also explores the underlying mechanisms and therapeutic potentials in these contexts. These insights aim to deepen our understanding of aging pathophysiology and the development of aging-related diseases.
Collapse
Affiliation(s)
- David Guo
- Cardiovascular Translational Research Center, Department of Internal Medicine, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ 85004, USA;
| | - Wuqiang Zhu
- Department of Cardiovascular Medicine, Physiology and Biomedical Engineering, Center for Regenerative Biotherapeutics, Mayo Clinic Arizona, Scottsdale, AZ 85259, USA;
| | - Hongyu Qiu
- Cardiovascular Translational Research Center, Department of Internal Medicine, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ 85004, USA;
- Clinical Translational Sciences (CTS) and Bio5 Institution, University of Arizona, Tucson, AZ 85721, USA
| |
Collapse
|
3
|
Cui X, Buonfiglio F, Pfeiffer N, Gericke A. Aging in Ocular Blood Vessels: Molecular Insights and the Role of Oxidative Stress. Biomedicines 2024; 12:817. [PMID: 38672172 PMCID: PMC11048681 DOI: 10.3390/biomedicines12040817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 04/01/2024] [Accepted: 04/05/2024] [Indexed: 04/28/2024] Open
Abstract
Acknowledged as a significant pathogenetic driver for numerous diseases, aging has become a focal point in addressing the profound changes associated with increasing human life expectancy, posing a critical concern for global public health. Emerging evidence suggests that factors influencing vascular aging extend their impact to choroidal and retinal blood vessels. The objective of this work is to provide a comprehensive overview of the impact of vascular aging on ocular blood vessels and related diseases. Additionally, this study aims to illuminate molecular insights contributing to vascular cell aging, with a particular emphasis on the choroid and retina. Moreover, innovative molecular targets operating within the domain of ocular vascular aging are presented and discussed.
Collapse
Affiliation(s)
- Xiuting Cui
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (F.B.); (N.P.)
| | | | | | - Adrian Gericke
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (F.B.); (N.P.)
| |
Collapse
|
4
|
Roubeix C, Nous C, Augustin S, Ronning KE, Mathis T, Blond F, Lagouge-Roussey P, Crespo-Garcia S, Sullivan PM, Gautier EL, Reichhart N, Sahel JA, Burns ME, Paques M, Sørensen TL, Strauss O, Guillonneau X, Delarasse C, Sennlaub F. Splenic monocytes drive pathogenic subretinal inflammation in age-related macular degeneration. J Neuroinflammation 2024; 21:22. [PMID: 38233865 PMCID: PMC10792815 DOI: 10.1186/s12974-024-03011-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 01/02/2024] [Indexed: 01/19/2024] Open
Abstract
Age-related macular degeneration (AMD) is invariably associated with the chronic accumulation of activated mononuclear phagocytes in the subretinal space. The mononuclear phagocytes are composed of microglial cells but also of monocyte-derived cells, which promote photoreceptor degeneration and choroidal neovascularization. Infiltrating blood monocytes can originate directly from bone marrow, but also from a splenic reservoir, where bone marrow monocytes develop into angiotensin II receptor (ATR1)+ splenic monocytes. The involvement of splenic monocytes in neurodegenerative diseases such as AMD is not well understood. Using acute inflammatory and well-phenotyped AMD models, we demonstrate that angiotensin II mobilizes ATR1+ splenic monocytes, which we show are defined by a transcriptional signature using single-cell RNA sequencing and differ functionally from bone marrow monocytes. Splenic monocytes participate in the chorio-retinal infiltration and their inhibition by ATR1 antagonist and splenectomy reduces the subretinal mononuclear phagocyte accumulation and pathological choroidal neovascularization formation. In aged AMD-risk ApoE2-expressing mice, a chronic AMD model, ATR1 antagonist and splenectomy also inhibit the chronic retinal inflammation and associated cone degeneration that characterizes these mice. Our observation of elevated levels of plasma angiotensin II in AMD patients, suggests that similar events take place in clinical disease and argue for the therapeutic potential of ATR1 antagonists to inhibit splenic monocytes for the treatment of blinding AMD.
Collapse
Affiliation(s)
- Christophe Roubeix
- Sorbonne Université, INSERM, CNRS, UMR_S 968, Institut de la Vision, 75012, Paris, France
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Experimental Ophthalmology, Department of Ophthalmology, Charitéplatz 1, 10117, Berlin, Germany
| | - Caroline Nous
- Sorbonne Université, INSERM, CNRS, UMR_S 968, Institut de la Vision, 75012, Paris, France
| | - Sébastien Augustin
- Sorbonne Université, INSERM, CNRS, UMR_S 968, Institut de la Vision, 75012, Paris, France
| | - Kaitryn E Ronning
- Sorbonne Université, INSERM, CNRS, UMR_S 968, Institut de la Vision, 75012, Paris, France
| | - Thibaud Mathis
- Service d'Ophtalmologie, Centre Hospitalier Universitaire de la Croix-Rousse, Hospices Civils de Lyon, Université Claude Bernard Lyon 1, 69004, Lyon, France
| | - Frédéric Blond
- Sorbonne Université, INSERM, CNRS, UMR_S 968, Institut de la Vision, 75012, Paris, France
| | | | - Sergio Crespo-Garcia
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Experimental Ophthalmology, Department of Ophthalmology, Charitéplatz 1, 10117, Berlin, Germany
| | - Patrick M Sullivan
- Department of Medicine, Centers for Aging and Geriatric Research Education and Clinical Center, Durham Veteran Affairs Medical Center, Duke University, Durham, NC, 27710, USA
| | - Emmanuel L Gautier
- Sorbonne Université, INSERM, UMR_S 1166, Hôpital de la Pitié-Salpêtrière, 75013, Paris, France
| | - Nadine Reichhart
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Experimental Ophthalmology, Department of Ophthalmology, Charitéplatz 1, 10117, Berlin, Germany
| | - José-Alain Sahel
- Sorbonne Université, INSERM, CNRS, UMR_S 968, Institut de la Vision, 75012, Paris, France
| | - Marie E Burns
- Center for Neuroscience, Department of Cell Biology and Human Anatomy, Department of Ophthalmology and Vision Science, University of California, Davis, CA, 95616, USA
| | - Michel Paques
- Sorbonne Université, INSERM, CNRS, UMR_S 968, Institut de la Vision, 75012, Paris, France
- Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, INSERM-DHOS Clinical Investigation Center 1423, Paris, France
| | - Torben Lykke Sørensen
- Clinical Eye Research Division, Department of Ophthalmology, Zealand University Hospital Roskilde, Roskilde, Denmark
- Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark
| | - Olaf Strauss
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Experimental Ophthalmology, Department of Ophthalmology, Charitéplatz 1, 10117, Berlin, Germany
| | - Xavier Guillonneau
- Sorbonne Université, INSERM, CNRS, UMR_S 968, Institut de la Vision, 75012, Paris, France
| | - Cécile Delarasse
- Sorbonne Université, INSERM, CNRS, UMR_S 968, Institut de la Vision, 75012, Paris, France.
| | - Florian Sennlaub
- Sorbonne Université, INSERM, CNRS, UMR_S 968, Institut de la Vision, 75012, Paris, France.
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Experimental Ophthalmology, Department of Ophthalmology, Charitéplatz 1, 10117, Berlin, Germany.
| |
Collapse
|
5
|
Tsioti I, Steiner BL, Escher P, Zinkernagel MS, Benz PM, Kokona D. Systemic Lipopolysaccharide Exposure Exacerbates Choroidal Neovascularization in Mice. Ocul Immunol Inflamm 2024; 32:19-30. [PMID: 36441988 DOI: 10.1080/09273948.2022.2147547] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 11/08/2022] [Indexed: 11/29/2022]
Abstract
This study aims to investigate the effect of a systemic lipopolysaccharide (LPS) stimulus in the course of laser-induced choroidal neovascularization (CNV) in C57BL/6 J mice. A group of CNV-subjected mice received 1 mg/kg LPS via the tail vein immediately after CNV induction. Mouse eyes were monitored in vivo with fluorescein angiography for 2 weeks. In situ hybridization and flow cytometry were performed in the retina at different time points. LPS led to increased fluorescein leakage 3 days after CNV, correlated with a large influx of monocyte-derived macrophages and increase of pro-inflammatory microglia/macrophages in the retina. Additionally, LPS enhanced Vegfα mRNA expression by Glul-expressing cells but not Aif1 positive microglia/macrophages in the laser lesion. These findings suggest that systemic LPS exposure has transient detrimental effects in the course of CNV through activation of microglia/macrophages to a pro-inflammatory phenotype and supports the important role of these cells in the CNV course.
Collapse
Affiliation(s)
- Ioanna Tsioti
- Department of Ophthalmology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Beatrice L Steiner
- Department of Ophthalmology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Pascal Escher
- Department of Ophthalmology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Martin S Zinkernagel
- Department of Ophthalmology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Peter M Benz
- Department of CardioMetabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Despina Kokona
- Department of Ophthalmology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department of BioMedical Research, University of Bern, Bern, Switzerland
| |
Collapse
|
6
|
Ping P, Guan L, Ning C, Liu Q, Zhao Y, Zhu X, Yang T, Fu S. WGCNA and molecular docking identify hub genes for cardiac aging. Front Cardiovasc Med 2023; 10:1146225. [PMID: 37180776 PMCID: PMC10172467 DOI: 10.3389/fcvm.2023.1146225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 04/10/2023] [Indexed: 05/16/2023] Open
Abstract
Background Cardiac aging and ageing-related cardiovascular diseases remain increase medical and social burden. Discovering the molecular mechanisms associated with cardiac aging is expected to provide new perspectives for delaying aging and related disease treatment. Methods The samples in GEO database were divided into older group and younger group based on age. Age-associated differentially expressed genes (DEGs) were identified by limma package. Gene modules significantly associated with age were mined using weighted gene co-expression network analysis (WGCNA). Protein-protein interaction networks (PPI) networks were developed using genes within modules, and topological analysis on the networks was performed to identify hub genes in cardiac aging. Pearson correlation was used to analyze the association among hub genes and immune and immune-related pathways. Molecular docking of hub genes and the anti-aging drug Sirolimus was performed to explore the potential role of hub genes in treating cardiac aging. Results We found a generally negative correlation between age and immunity, with a significant negative correlation between age and b_cell_receptor_signaling_pathway, fc_gamma_r_mediated_phagocytosis, chemokine signaling pathway, t-cell receptor signaling pathway, toll_like_receptor_signaling_pathway, and jak_stat_signaling_pathway, respectively. Finally, 10 cardiac aging-related hub genes including LCP2, PTPRC, RAC2, CD48, CD68, CCR2, CCL2, IL10, CCL5 and IGF1 were identified. 10-hub genes were closely associated with age and immune-related pathways. There was a strong binding interaction between Sirolimus-CCR2. CCR2 may be a key target for Sirolimus in the treatment of cardiac aging. Conclusion The 10 hub genes may be potential therapeutic targets for cardiac aging, and our study provided new ideas for the treatment of cardiac aging.
Collapse
Affiliation(s)
- Ping Ping
- General Station for Drug and Instrument Supervision and Control, Joint Logistic Support Force of Chinese People's Liberation Army, Beijing, China
| | - Lixun Guan
- Hematology Department, Hainan Hospital of Chinese People's Liberation Army General Hospital, Sanya, China
| | - Chaoxue Ning
- Central Laboratory, Hainan Hospital of Chinese People's Liberation Army General Hospital, Sanya, China
| | - Qiong Liu
- Medical Care Center, Hainan Hospital of Chinese People's Liberation Army General Hospital, Sanya, China
| | - Yali Zhao
- Central Laboratory, Hainan Hospital of Chinese People's Liberation Army General Hospital, Sanya, China
| | - Xiang Zhu
- Department of Infectious Disease, Army No.82 Group Military Hospital, Baoding, China
| | - Ting Yang
- Central Laboratory, Hainan Hospital of Chinese People's Liberation Army General Hospital, Sanya, China
| | - Shihui Fu
- Department of Cardiology, Hainan Hospital of Chinese People's Liberation Army General Hospital, Sanya, China
- Department of Geriatric Cardiology, Chinese People's Liberation Army General Hospital, Beijing, China
| |
Collapse
|
7
|
Mokhtari K, Peymani M, Rashidi M, Hushmandi K, Ghaedi K, Taheriazam A, Hashemi M. Colon cancer transcriptome. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2023; 180-181:49-82. [PMID: 37059270 DOI: 10.1016/j.pbiomolbio.2023.04.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/31/2023] [Accepted: 04/06/2023] [Indexed: 04/16/2023]
Abstract
Over the last four decades, methodological innovations have continuously changed transcriptome profiling. It is now feasible to sequence and quantify the transcriptional outputs of individual cells or thousands of samples using RNA sequencing (RNA-seq). These transcriptomes serve as a connection between cellular behaviors and their underlying molecular mechanisms, such as mutations. This relationship, in the context of cancer, provides a chance to unravel tumor complexity and heterogeneity and uncover novel biomarkers or treatment options. Since colon cancer is one of the most frequent malignancies, its prognosis and diagnosis seem to be critical. The transcriptome technology is developing for an earlier and more accurate diagnosis of cancer which can provide better protectivity and prognostic utility to medical teams and patients. A transcriptome is a whole set of expressed coding and non-coding RNAs in an individual or cell population. The cancer transcriptome includes RNA-based changes. The combined genome and transcriptome of a patient may provide a comprehensive picture of their cancer, and this information is beginning to affect treatment decision-making in real-time. A full assessment of the transcriptome of colon (colorectal) cancer has been assessed in this review paper based on risk factors such as age, obesity, gender, alcohol use, race, and also different stages of cancer, as well as non-coding RNAs like circRNAs, miRNAs, lncRNAs, and siRNAs. Similarly, they have been examined independently in the transcriptome study of colon cancer.
Collapse
Affiliation(s)
- Khatere Mokhtari
- Department of Modern Biology, ACECR Institute of Higher Education (Isfahan Branch), Isfahan, Iran
| | - Maryam Peymani
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran.
| | - Mohsen Rashidi
- Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, 4815733971, Iran; The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, 4815733971, Iran
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Kamran Ghaedi
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran.
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
8
|
Palazzo I, Todd LJ, Hoang TV, Reh TA, Blackshaw S, Fischer AJ. NFkB-signaling promotes glial reactivity and suppresses Müller glia-mediated neuron regeneration in the mammalian retina. Glia 2022; 70:1380-1401. [PMID: 35388544 DOI: 10.1002/glia.24181] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 12/25/2022]
Abstract
Müller glia (MG) in mammalian retinas are incapable of regenerating neurons after damage, whereas the MG in lower vertebrates regenerate functional neurons. Identification of cell signaling pathways and gene regulatory networks that regulate MG-mediated regeneration is key to harnessing the regenerative potential of MG. Here, we study how NFkB-signaling influences glial responses to damage and reprogramming of MG into neurons in the rodent retina. We find activation of NFkB and dynamic expression of NFkB-associated genes in MG after damage, however damage-induced NFkB activation is inhibited by microglia ablation. Knockout of NFkB in MG suppressed the accumulation of immune cells after damage. Inhibition of NFkB following NMDA-damage significantly enhanced the reprogramming of Ascl1-overexpressing MG into neuron-like cells. scRNA-seq of retinal glia following inhibition of NFkB reveals coordination with signaling via TGFβ2 and suppression of NFI and Id transcription factors. Inhibition of Smad3 signal transducer or Id transcription factors increased numbers of neuron-like cells produced by Ascl1-overexpressing MG. We conclude that NFkB is a key signaling hub that is activated in MG after damage, mediates the accumulation of immune cells, and suppresses the neurogenic potential of MG.
Collapse
Affiliation(s)
- Isabella Palazzo
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Levi J Todd
- Department of Biological Structure, College of Medicine, University of Washington, Seattle, Washington, USA
| | - Thanh V Hoang
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Thomas A Reh
- Department of Biological Structure, College of Medicine, University of Washington, Seattle, Washington, USA
| | - Seth Blackshaw
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Andy J Fischer
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
9
|
Martínez-Alberquilla I, Gasull X, Pérez-Luna P, Seco-Mera R, Ruiz-Alcocer J, Crooke A. Neutrophils and neutrophil extracellular trap components: Emerging biomarkers and therapeutic targets for age-related eye diseases. Ageing Res Rev 2022; 74:101553. [PMID: 34971794 DOI: 10.1016/j.arr.2021.101553] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 11/17/2021] [Accepted: 12/23/2021] [Indexed: 12/14/2022]
Abstract
Age-related eye diseases, including dry eye, glaucoma, age-related macular degeneration, and diabetic retinopathy, represent a major global health issue based on their increasing prevalence and disabling action. Unraveling the molecular mechanisms underlying these diseases will provide novel opportunities to reduce the burden of age-related eye diseases and improve eye health, contributing to sustainable development goals achievement. The impairment of neutrophil extracellular traps formation/degradation processes seems to be one of these mechanisms. These traps formed by a meshwork of DNA and neutrophil cytosolic granule proteins may exacerbate the inflammatory response promoting chronic inflammation, a pivotal cause of age-related diseases. In this review, we describe current findings that suggest the role of neutrophils and their traps in the pathogenesis of the above-mentioned age-related eye diseases. Furthermore, we discuss why these cells and their constituents could be biomarkers and therapeutic targets for dry eye, glaucoma, age-related macular degeneration, and diabetic retinopathy. We also examine the therapeutic potential of some neutrophil function modulators and provide several recommendations for future research in age-related eye diseases.
Collapse
Affiliation(s)
- Irene Martínez-Alberquilla
- Department of Optometry and Vision, Faculty of Optics and Optometry, Universidad Complutense de Madrid, Madrid, Spain; Clinical and Experimental Eye Research Group, UCM 971009, Faculty of Optics and Optometry, Universidad Complutense de Madrid, Madrid, Spain
| | - Xavier Gasull
- Neurophysiology Laboratory, Department of Biomedicine, Medical School, Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Patricia Pérez-Luna
- Department of Biochemistry and Molecular Biology, Faculty of Optics and Optometry, Universidad Complutense de Madrid, Madrid, Spain
| | - Rubén Seco-Mera
- Department of Biochemistry and Molecular Biology, Faculty of Optics and Optometry, Universidad Complutense de Madrid, Madrid, Spain
| | - Javier Ruiz-Alcocer
- Department of Optometry and Vision, Faculty of Optics and Optometry, Universidad Complutense de Madrid, Madrid, Spain; Clinical and Experimental Eye Research Group, UCM 971009, Faculty of Optics and Optometry, Universidad Complutense de Madrid, Madrid, Spain
| | - Almudena Crooke
- Department of Biochemistry and Molecular Biology, Faculty of Optics and Optometry, Universidad Complutense de Madrid, Madrid, Spain; Clinical and Experimental Eye Research Group, UCM 971009, Faculty of Optics and Optometry, Universidad Complutense de Madrid, Madrid, Spain.
| |
Collapse
|
10
|
Touhami S, Béguier F, Yang T, Augustin S, Roubeix C, Blond F, Conart JB, Sahel JA, Bodaghi B, Delarasse C, Guillonneau X, Sennlaub F. Hypoxia Inhibits Subretinal Inflammation Resolution Thrombospondin-1 Dependently. Int J Mol Sci 2022; 23:681. [PMID: 35054863 PMCID: PMC8775350 DOI: 10.3390/ijms23020681] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 01/02/2022] [Accepted: 01/04/2022] [Indexed: 12/10/2022] Open
Abstract
Hypoxia is potentially one of the essential triggers in the pathogenesis of wet age-related macular degeneration (wetAMD), characterized by choroidal neovascularization (CNV) which is driven by the accumulation of subretinal mononuclear phagocytes (MP) that include monocyte-derived cells. Here we show that systemic hypoxia (10% O2) increased subretinal MP infiltration and inhibited inflammation resolution after laser-induced subretinal injury in vivo. Accordingly, hypoxic (2% O2) human monocytes (Mo) resisted elimination by RPE cells in co-culture. In Mos from hypoxic mice, Thrombospondin 1 mRNA (Thbs1) was most downregulated compared to normoxic animals and hypoxia repressed Thbs-1 expression in human monocytes in vitro. Hypoxic ambient air inhibited MP clearance during the resolution phase of laser-injury in wildtype animals, but had no effect on the exaggerated subretinal MP infiltration observed in normoxic Thbs1-/--mice. Recombinant Thrombospondin 1 protein (TSP-1) completely reversed the pathogenic effect of hypoxia in Thbs1-/--mice, and accelerated inflammation resolution and inhibited CNV in wildtype mice. Together, our results demonstrate that systemic hypoxia disturbs TSP-1-dependent subretinal immune suppression and promotes pathogenic subretinal inflammation and can be therapeutically countered by local recombinant TSP-1.
Collapse
Affiliation(s)
- Sara Touhami
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, 75012 Paris, France; (S.T.); (F.B.); (T.Y.); (S.A.); (C.R.); (F.B.); (J.B.C.); (J.A.S.); (C.D.); (X.G.)
- Ophthalmology Department, Pitié Salpêtrière University Hospital, Sorbonne Université, AP-HP, 75013 Paris, France;
| | - Fanny Béguier
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, 75012 Paris, France; (S.T.); (F.B.); (T.Y.); (S.A.); (C.R.); (F.B.); (J.B.C.); (J.A.S.); (C.D.); (X.G.)
| | - Tianxiang Yang
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, 75012 Paris, France; (S.T.); (F.B.); (T.Y.); (S.A.); (C.R.); (F.B.); (J.B.C.); (J.A.S.); (C.D.); (X.G.)
| | - Sébastien Augustin
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, 75012 Paris, France; (S.T.); (F.B.); (T.Y.); (S.A.); (C.R.); (F.B.); (J.B.C.); (J.A.S.); (C.D.); (X.G.)
| | - Christophe Roubeix
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, 75012 Paris, France; (S.T.); (F.B.); (T.Y.); (S.A.); (C.R.); (F.B.); (J.B.C.); (J.A.S.); (C.D.); (X.G.)
| | - Frederic Blond
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, 75012 Paris, France; (S.T.); (F.B.); (T.Y.); (S.A.); (C.R.); (F.B.); (J.B.C.); (J.A.S.); (C.D.); (X.G.)
| | - Jean Baptiste Conart
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, 75012 Paris, France; (S.T.); (F.B.); (T.Y.); (S.A.); (C.R.); (F.B.); (J.B.C.); (J.A.S.); (C.D.); (X.G.)
- Department of Ophthalmology, University Hospital, 54000 Nancy, France
| | - José Alain Sahel
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, 75012 Paris, France; (S.T.); (F.B.); (T.Y.); (S.A.); (C.R.); (F.B.); (J.B.C.); (J.A.S.); (C.D.); (X.G.)
- CHNO des Quinze-Vingts, INSERM-DGOS CIC 1423, 75012 Paris, France
| | - Bahram Bodaghi
- Ophthalmology Department, Pitié Salpêtrière University Hospital, Sorbonne Université, AP-HP, 75013 Paris, France;
| | - Cécile Delarasse
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, 75012 Paris, France; (S.T.); (F.B.); (T.Y.); (S.A.); (C.R.); (F.B.); (J.B.C.); (J.A.S.); (C.D.); (X.G.)
| | - Xavier Guillonneau
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, 75012 Paris, France; (S.T.); (F.B.); (T.Y.); (S.A.); (C.R.); (F.B.); (J.B.C.); (J.A.S.); (C.D.); (X.G.)
| | - Florian Sennlaub
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, 75012 Paris, France; (S.T.); (F.B.); (T.Y.); (S.A.); (C.R.); (F.B.); (J.B.C.); (J.A.S.); (C.D.); (X.G.)
| |
Collapse
|
11
|
Immunosenescence in Choroidal Neovascularization (CNV)-Transcriptional Profiling of Naïve and CNV-Associated Retinal Myeloid Cells during Aging. Int J Mol Sci 2021; 22:ijms222413318. [PMID: 34948115 PMCID: PMC8707893 DOI: 10.3390/ijms222413318] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/06/2021] [Accepted: 12/08/2021] [Indexed: 02/08/2023] Open
Abstract
Immunosenescence is considered a possible factor in the development of age-related macular degeneration and choroidal neovascularization (CNV). However, age-related changes of myeloid cells (MCs), such as microglia and macrophages, in the healthy retina or during CNV formation are ill-defined. In this study, Cx3cr1-positive MCs were isolated by fluorescence-activated cell sorting from six-week (young) and two-year-old (old) Cx3cr1GFP/+ mice, both during physiological aging and laser-induced CNV development. High-throughput RNA-sequencing was performed to define the age-dependent transcriptional differences in MCs during physiological aging and CNV development, complemented by immunohistochemical characterization and the quantification of MCs, as well as CNV size measurements. These analyses revealed that myeloid cells change their transcriptional profile during both aging and CNV development. In the steady state, senescent MCs demonstrated an upregulation of factors contributing to cell proliferation and chemotaxis, such as Cxcl13 and Cxcl14, as well as the downregulation of microglial signature genes. During CNV formation, aged myeloid cells revealed a significant upregulation of angiogenic factors such as Arg1 and Lrg1 concomitant with significantly enlarged CNV and an increased accumulation of MCs in aged mice in comparison to young mice. Future studies need to clarify whether this observation is an epiphenomenon or a causal relationship to determine the role of immunosenescence in CNV formation.
Collapse
|
12
|
Lu ZG, May A, Dinh B, Lin V, Su F, Tran C, Adivikolanu H, Ehlen R, Che B, Wang ZH, Shaw DH, Borooah S, Shaw PX. The interplay of oxidative stress and ARMS2-HTRA1 genetic risk in neovascular AMD. ACTA ACUST UNITED AC 2021; 5. [PMID: 34017939 PMCID: PMC8133762 DOI: 10.20517/2574-1209.2020.48] [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] [Indexed: 12/27/2022]
Abstract
Age-related macular degeneration (AMD) is the leading cause of vision loss in adults over 60 years old globally. There are two forms of advanced AMD: “dry” and “wet”. Dry AMD is characterized by geographic atrophy of the retinal pigment epithelium and overlying photoreceptors in the macular region; whereas wet AMD is characterized by vascular penetrance from the choroid into the retina, known as choroidal neovascularization (CNV). Both phenotypes eventually lead to loss of central vision. The pathogenesis of AMD involves the interplay of genetic polymorphisms and environmental risk factors, many of which elevate retinal oxidative stress. Excess reactive oxygen species react with cellular macromolecules, forming oxidation-modified byproducts that elicit chronic inflammation and promote CNV. Additionally, genome-wide association studies have identified several genetic variants in the age-related maculopathy susceptibility 2/high-temperature requirement A serine peptidase 1 (ARMS2-HTRA1) locus associated with the progression of late-stage AMD, especially the wet subtype. In this review, we will focus on the interplay of oxidative stress and HTRA1 in drusen deposition, chronic inflammation, and chronic angiogenesis. We aim to present a multifactorial model of wet AMD progression, supporting HTRA1 as a novel therapeutic target upstream of vascular endothelial growth factor (VEGF), the conventional target in AMD therapeutics. By inhibiting HTRA1’s proteolytic activity, we can reduce pro-angiogenic signaling and prevent proteolytic breakdown of the blood-retina barrier. The anti-HTRA1 approach offers a promising alternative treatment option to wet AMD, complementary to anti-VEGF therapy.
Collapse
Affiliation(s)
- Zhi-Gang Lu
- Department of Neurology, First People's Hospital of Jingmen, Jingchu University of Technology, Jingmen 448000, Hubei, China.,Viterbi Family Department of Ophthalmology and Shiley Eye Institute, University of California, San Diego, La Jolla, CA 92093, USA.,Altman Clinical and Translational Research Institute, University of California, San Diego, La Jolla, CA 92093, USA
| | - Adam May
- Viterbi Family Department of Ophthalmology and Shiley Eye Institute, University of California, San Diego, La Jolla, CA 92093, USA.,Altman Clinical and Translational Research Institute, University of California, San Diego, La Jolla, CA 92093, USA
| | - Brian Dinh
- Viterbi Family Department of Ophthalmology and Shiley Eye Institute, University of California, San Diego, La Jolla, CA 92093, USA.,Altman Clinical and Translational Research Institute, University of California, San Diego, La Jolla, CA 92093, USA
| | - Victor Lin
- Viterbi Family Department of Ophthalmology and Shiley Eye Institute, University of California, San Diego, La Jolla, CA 92093, USA.,Altman Clinical and Translational Research Institute, University of California, San Diego, La Jolla, CA 92093, USA
| | - Fei Su
- Viterbi Family Department of Ophthalmology and Shiley Eye Institute, University of California, San Diego, La Jolla, CA 92093, USA.,Altman Clinical and Translational Research Institute, University of California, San Diego, La Jolla, CA 92093, USA
| | - Christina Tran
- Viterbi Family Department of Ophthalmology and Shiley Eye Institute, University of California, San Diego, La Jolla, CA 92093, USA.,Altman Clinical and Translational Research Institute, University of California, San Diego, La Jolla, CA 92093, USA
| | - Harini Adivikolanu
- Viterbi Family Department of Ophthalmology and Shiley Eye Institute, University of California, San Diego, La Jolla, CA 92093, USA.,Altman Clinical and Translational Research Institute, University of California, San Diego, La Jolla, CA 92093, USA
| | - Rachael Ehlen
- Viterbi Family Department of Ophthalmology and Shiley Eye Institute, University of California, San Diego, La Jolla, CA 92093, USA.,Altman Clinical and Translational Research Institute, University of California, San Diego, La Jolla, CA 92093, USA
| | - Briana Che
- Viterbi Family Department of Ophthalmology and Shiley Eye Institute, University of California, San Diego, La Jolla, CA 92093, USA.,Altman Clinical and Translational Research Institute, University of California, San Diego, La Jolla, CA 92093, USA
| | - Zhi-Hao Wang
- Viterbi Family Department of Ophthalmology and Shiley Eye Institute, University of California, San Diego, La Jolla, CA 92093, USA.,Altman Clinical and Translational Research Institute, University of California, San Diego, La Jolla, CA 92093, USA
| | - Daniel H Shaw
- Viterbi Family Department of Ophthalmology and Shiley Eye Institute, University of California, San Diego, La Jolla, CA 92093, USA.,Altman Clinical and Translational Research Institute, University of California, San Diego, La Jolla, CA 92093, USA.,Westview High School, San Diego, CA 92131, USA
| | - Shyamanga Borooah
- Viterbi Family Department of Ophthalmology and Shiley Eye Institute, University of California, San Diego, La Jolla, CA 92093, USA
| | - Peter X Shaw
- Viterbi Family Department of Ophthalmology and Shiley Eye Institute, University of California, San Diego, La Jolla, CA 92093, USA.,Altman Clinical and Translational Research Institute, University of California, San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
13
|
Krogh Nielsen M, Subhi Y, Molbech CR, Falk MK, Nissen MH, Sørensen TL. Chemokine Profile and the Alterations in CCR5-CCL5 Axis in Geographic Atrophy Secondary to Age-Related Macular Degeneration. Invest Ophthalmol Vis Sci 2020; 61:28. [PMID: 32324857 PMCID: PMC7401724 DOI: 10.1167/iovs.61.4.28] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Purpose Geographic atrophy (GA) secondary to age-related macular degeneration (AMD) is a progressive disease with no treatment option. Previous studies show chemokine-mediated recruitment of immune cells in the retina, and therefore we investigated systemic levels of chemokines and chemokine receptors in patients with GA. Methods This observational prospective study was conducted at a single center. We included 122 participants with no immune disease: 41 participants with GA and no choroidal neovascularization, 51 patients with neovascular AMD, and 30 healthy control individuals. Flow cytometric analysis was used to detect expression level of C-C chemokine receptor (CCR)1, CCR2, CCR3, CCR5, and C-X-C motif chemokine receptor (CXCR)3 on peripheral blood mononuclear cells (CD14+ monocytes, CD4+ T cells, CD8+ T cells). Plasma levels of C-C motif ligand (CCL)11, C-X-C motif chemokine (CXCL)10, and CCL5 were measured by specific immunoassays. Enlargement rate of GA lesion was measured from autofluorescence images. Results Participants with GA have a specific chemokine profile with a higher expression of CCR5 than healthy controls in peripheral blood mononuclear cells, and a higher plasma levels of CCL-5. Further, GA was associated with higher monocytic expression of CCR2 than in neovascular AMD. We found that a high expression level of CCR5 on CD8+ T cells was associated with slower enlargement rate of atrophic lesion. Conclusions The study showed an association between systemic chemokine profile and GA formation. Further studies are needed to fully elucidate the possible role of systemic chemokine regulation in mediating pathogenesis of GA.
Collapse
|
14
|
Beguier F, Housset M, Roubeix C, Augustin S, Zagar Y, Nous C, Mathis T, Eandi C, Benchaboune M, Drame-Maigné A, Carpentier W, Chardonnet S, Touhami S, Blot G, Conart JB, Charles-Messance H, Potey A, Girmens JF, Paques M, Blond F, Leveillard T, Koertvely E, Roger JE, Sahel JA, Sapieha P, Delarasse C, Guillonneau X, Sennlaub F. The 10q26 Risk Haplotype of Age-Related Macular Degeneration Aggravates Subretinal Inflammation by Impairing Monocyte Elimination. Immunity 2020; 53:429-441.e8. [PMID: 32814029 DOI: 10.1016/j.immuni.2020.07.021] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 03/06/2020] [Accepted: 07/24/2020] [Indexed: 02/08/2023]
Abstract
A minor haplotype of the 10q26 locus conveys the strongest genetic risk for age-related macular degeneration (AMD). Here, we examined the mechanisms underlying this susceptibility. We found that monocytes from homozygous carriers of the 10q26 AMD-risk haplotype expressed high amounts of the serine peptidase HTRA1, and HTRA1 located to mononuclear phagocytes (MPs) in eyes of non-carriers with AMD. HTRA1 induced the persistence of monocytes in the subretinal space and exacerbated pathogenic inflammation by hydrolyzing thrombospondin 1 (TSP1), which separated the two CD47-binding sites within TSP1 that are necessary for efficient CD47 activation. This HTRA1-induced inhibition of CD47 signaling induced the expression of pro-inflammatory osteopontin (OPN). OPN expression increased in early monocyte-derived macrophages in 10q26 risk carriers. In models of subretinal inflammation and AMD, OPN deletion or pharmacological inhibition reversed HTRA1-induced pathogenic MP persistence. Our findings argue for the therapeutic potential of CD47 agonists and OPN inhibitors for the treatment of AMD.
Collapse
Affiliation(s)
- Fanny Beguier
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France
| | - Michael Housset
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France
| | - Christophe Roubeix
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France
| | - Sebastien Augustin
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France
| | - Yvrick Zagar
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France
| | - Caroline Nous
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France
| | - Thibaud Mathis
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France
| | - Chiara Eandi
- University of Torino, Department of Surgical Science, Torino, Italy
| | - Mustapha Benchaboune
- CHNO des Quinze-Vingts, DHU Sight Restore, INSERM-DGOS CIC 1423, 28 rue de Charenton, F-75012 Paris, France
| | - Adèle Drame-Maigné
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France
| | - Wassila Carpentier
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France
| | - Solenne Chardonnet
- Sorbonne Université, INSERM, UMS 37 PASS, Plateforme Post-génomique de la Pitié-Salpêtrière, P3S, F-75013 Paris, France
| | - Sara Touhami
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France
| | - Guillaume Blot
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France
| | - Jean Baptiste Conart
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France
| | - Hugo Charles-Messance
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France
| | - Anaïs Potey
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France
| | - Jean-François Girmens
- CHNO des Quinze-Vingts, DHU Sight Restore, INSERM-DGOS CIC 1423, 28 rue de Charenton, F-75012 Paris, France
| | - Michel Paques
- CHNO des Quinze-Vingts, DHU Sight Restore, INSERM-DGOS CIC 1423, 28 rue de Charenton, F-75012 Paris, France
| | - Fréderic Blond
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France
| | - Thierry Leveillard
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France
| | - Elod Koertvely
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, 124 Grenzacherstrasse, 4070, Basel, Switzerland
| | - Jerome E Roger
- Paris-Saclay Institute of Neuroscience, CERTO-Retina France, CNRS, Univ Paris Sud, Université Paris-Saclay, F-91405 Orsay
| | - José-Alain Sahel
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France; CHNO des Quinze-Vingts, DHU Sight Restore, INSERM-DGOS CIC 1423, 28 rue de Charenton, F-75012 Paris, France
| | - Przemyslaw Sapieha
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Quebec, Canada
| | - Cécile Delarasse
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France
| | - Xavier Guillonneau
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France
| | - Florian Sennlaub
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France.
| |
Collapse
|
15
|
Yu C, Roubeix C, Sennlaub F, Saban DR. Microglia versus Monocytes: Distinct Roles in Degenerative Diseases of the Retina. Trends Neurosci 2020; 43:433-449. [PMID: 32459994 DOI: 10.1016/j.tins.2020.03.012] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 02/27/2020] [Accepted: 03/25/2020] [Indexed: 01/22/2023]
Abstract
Unlike in the healthy mammalian retina, macrophages in retinal degenerative states are not solely comprised of microglia but may include monocyte-derived recruits. Recent studies have applied transgenics, lineage-tracing, and transcriptomics to help decipher the distinct roles of these two cell types in the diseasesettings of inherited retinal degenerations and age-related macular degeneration.Literature discussed here focuses on the ectopic presence of both macrophage types in the extracellular site surrounding the outer aspect ofphotoreceptor cells (i.e.,the subretinal space), which is crucially involved in the pathobiology. From these studies we propose a working model in which perturbed photoreceptor states cause microglial dominant migration to the subretinal space as a protective response, whereas the abundant presence ofmonocyte-derived cells there instead drives and accelerates pathology. The latter, we propose, is underpinned by specific genetic and nongenetic determinants that lead to a maladaptive macrophage state.
Collapse
Affiliation(s)
- Chen Yu
- Department of Ophthalmology, Duke University, Durham, NC 27710, USA
| | - Christophe Roubeix
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, F-75012 Paris, France
| | - Florian Sennlaub
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, F-75012 Paris, France.
| | - Daniel R Saban
- Department of Ophthalmology, Duke University, Durham, NC 27710, USA; Department of Immunology, Duke University,Durham, NC 27710, USA.
| |
Collapse
|
16
|
Will-Orrego A, Qiu Y, Fassbender ES, Shen S, Aranda J, Kotagiri N, Maker M, Liao SM, Jaffee BD, Poor SH. Amount of Mononuclear Phagocyte Infiltrate Does Not Predict Area of Experimental Choroidal Neovascularization (CNV). J Ocul Pharmacol Ther 2019; 34:489-499. [PMID: 30188257 PMCID: PMC6152860 DOI: 10.1089/jop.2017.0131] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Purpose: Mononuclear phagocytes (MNPs) are present in neovascular age-related macular degeneration (nv AMD) which is also called choroidal neovascularization (CNV). The number and phenotype of the MNPs depend upon the local environment in the CNV and effect of nv AMD therapy. We investigated ocular cell infiltration and conditions that modulate angiogenesis in a laser-induced mouse CNV model. Methods: We developed assays to quantify MNPs in our established mouse CNV model. One MNP assay quantified the number of subretinal cells peripheral to the CNV lesions. A second assay semiquantitatively assesses the number of MNPs localized to the CNV lesion. We used these assays to measure the effect of toll-like receptor-2 (TLR-2) activation, anti-vascular endothelial growth factor (VEGF) therapy, and chemokine (C-C motif) ligand 2 (Ccl2) genetic deletion on MNP infiltration after laser injury. Results: Laser injury induced blood vessel growth and infiltration of MNPs. Systemic administration of a TLR-2 activating peptide increased laser-induced CNV area, MNP cell numbers, and MNP density over the CNV lesions. Systemic administration of a VEGF antibody reduced CNV area, while Ccl2 genetic deletion increased CNV area. Despite the change in amount of angiogenesis, MNP infiltration was, surprisingly, unchanged in these 2 conditions. Conclusions: MNP quantification provides biological insights for candidate AMD therapies. The number of infiltrating MNP cells does not correlate with the amount of laser-induced CNV area.
Collapse
Affiliation(s)
- Adrian Will-Orrego
- Department of Ophthalmology, Novartis Institutes for Biomedical Research , Cambridge, Massachusetts
| | - Yubin Qiu
- Department of Ophthalmology, Novartis Institutes for Biomedical Research , Cambridge, Massachusetts
| | - Elizabeth S Fassbender
- Department of Ophthalmology, Novartis Institutes for Biomedical Research , Cambridge, Massachusetts
| | - Siyuan Shen
- Department of Ophthalmology, Novartis Institutes for Biomedical Research , Cambridge, Massachusetts
| | - Jorge Aranda
- Department of Ophthalmology, Novartis Institutes for Biomedical Research , Cambridge, Massachusetts
| | - Namrata Kotagiri
- Department of Ophthalmology, Novartis Institutes for Biomedical Research , Cambridge, Massachusetts
| | - Michael Maker
- Department of Ophthalmology, Novartis Institutes for Biomedical Research , Cambridge, Massachusetts
| | - Sha-Mei Liao
- Department of Ophthalmology, Novartis Institutes for Biomedical Research , Cambridge, Massachusetts
| | - Bruce D Jaffee
- Department of Ophthalmology, Novartis Institutes for Biomedical Research , Cambridge, Massachusetts
| | - Stephen H Poor
- Department of Ophthalmology, Novartis Institutes for Biomedical Research , Cambridge, Massachusetts
| |
Collapse
|
17
|
Transcriptome analysis identifies a robust gene expression program in the mouse intestinal epithelium on aging. Sci Rep 2019; 9:10410. [PMID: 31320724 PMCID: PMC6639340 DOI: 10.1038/s41598-019-46966-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 07/03/2019] [Indexed: 12/14/2022] Open
Abstract
The intestinal epithelium undergoes constant regeneration driven by intestinal stem cells. How old age affects the transcriptome in this highly dynamic tissue is an important, but poorly explored question. Using transcriptomics on sorted intestinal stem cells and adult enterocytes, we identified candidate genes, which change expression on aging. Further validation of these on intestinal epithelium of multiple middle-aged versus old-aged mice highlighted the consistent up-regulation of the expression of the gene encoding chemokine receptor Ccr2, a mediator of inflammation and several disease processes. We observed also increased expression of Strc, coding for stereocilin, and dramatically decreased expression of Rps4l, coding for a ribosome subunit. Ccr2 and Rps4l are located close to the telomeric regions of chromosome 9 and 6, respectively. As only few genes were differentially expressed and we did not observe significant protein level changes of identified ageing markers, our analysis highlights the overall robustness of murine intestinal epithelium gene expression to old age.
Collapse
|
18
|
Akhtar-Schäfer I, Wang L, Krohne TU, Xu H, Langmann T. Modulation of three key innate immune pathways for the most common retinal degenerative diseases. EMBO Mol Med 2019; 10:emmm.201708259. [PMID: 30224384 PMCID: PMC6180304 DOI: 10.15252/emmm.201708259] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
This review highlights the role of three key immune pathways in the pathophysiology of major retinal degenerative diseases including diabetic retinopathy, age‐related macular degeneration, and rare retinal dystrophies. We first discuss the mechanisms how loss of retinal homeostasis evokes an unbalanced retinal immune reaction involving responses of local microglia and recruited macrophages, activity of the alternative complement system, and inflammasome assembly in the retinal pigment epithelium. Presenting these key mechanisms as complementary targets, we specifically emphasize the concept of immunomodulation as potential treatment strategy to prevent or delay vision loss. Promising molecules are ligands for phagocyte receptors, specific inhibitors of complement activation products, and inflammasome inhibitors. We comprehensively summarize the scientific evidence for this strategy from preclinical animal models, human ocular tissue analyses, and clinical trials evolving in the last few years.
Collapse
Affiliation(s)
- Isha Akhtar-Schäfer
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, University of Cologne, Cologne, Germany
| | - Luping Wang
- Department of Ophthalmology, University of Bonn, Bonn, Germany
| | - Tim U Krohne
- Department of Ophthalmology, University of Bonn, Bonn, Germany
| | - Heping Xu
- Centre for Experimental Medicine, The Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Sciences, Queen's University Belfast, Belfast, UK
| | - Thomas Langmann
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, University of Cologne, Cologne, Germany .,Center for Molecular Medicine, University of Cologne, Cologne, Germany
| |
Collapse
|
19
|
Murinello S, Usui Y, Sakimoto S, Kitano M, Aguilar E, Friedlander HM, Schricker A, Wittgrove C, Wakabayashi Y, Dorrell MI, Westenskow PD, Friedlander M. miR-30a-5p inhibition promotes interaction of Fas + endothelial cells and FasL + microglia to decrease pathological neovascularization and promote physiological angiogenesis. Glia 2018; 67:332-344. [PMID: 30484883 DOI: 10.1002/glia.23543] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 09/05/2018] [Accepted: 09/05/2018] [Indexed: 12/18/2022]
Abstract
Ischemia-induced angiogenesis contributes to various neuronal and retinal diseases, and often results in neurodegeneration and visual impairment. Current treatments involve the use of anti-VEGF agents but are not successful in all cases. In this study we determined that miR-30a-5p is another important mediator of retinal angiogenesis. Using a rodent model of ischemic retinopathy, we show that inhibiting miR-30a-5p reduces neovascularization and promotes tissue repair, through modulation of microglial and endothelial cell cross-talk. miR-30a-5p inhibition results in increased expression of the death receptor Fas and CCL2, to decrease endothelial cell survival and promote microglial migration and phagocytic function in focal regions of ischemic injury. Our data suggest that miR-30a-5p inhibition accelerates tissue repair by enhancing FasL-Fas crosstalk between microglia and endothelial cells, to promote endothelial cell apoptosis and removal of dead endothelial cells. Finally, we found that miR-30a levels were increased in the vitreous of patients with proliferative diabetic retinopathy. Our study identifies a role for miR-30a in the pathogenesis of neovascular retinal disease by modulating microglial and endothelial cell function, and suggests it may be a therapeutic target to treat ischemia-mediated conditions.
Collapse
Affiliation(s)
- Salome Murinello
- Department of Molecular Medicine, The Scripps Research Institute, California
| | - Yoshihiko Usui
- Department of Ophthalmology, Tokyo Medical University, Tokyo, Japan
| | - Susumu Sakimoto
- Department of Molecular Medicine, The Scripps Research Institute, California
| | - Maki Kitano
- Department of Molecular Medicine, The Scripps Research Institute, California
| | - Edith Aguilar
- Department of Molecular Medicine, The Scripps Research Institute, California
| | - H Maura Friedlander
- Department of Molecular Medicine, The Scripps Research Institute, California
| | - Amelia Schricker
- Department of Molecular Medicine, The Scripps Research Institute, California
| | - Carli Wittgrove
- Department of Molecular Medicine, The Scripps Research Institute, California
| | | | - Michael I Dorrell
- Department of Molecular Medicine, The Scripps Research Institute, California.,The Lowy Medical Research Institute, California.,Department of Biology, Point Loma Nazarene University, San Diego, California
| | - Peter D Westenskow
- Department of Molecular Medicine, The Scripps Research Institute, California
| | - Martin Friedlander
- Department of Molecular Medicine, The Scripps Research Institute, California.,The Lowy Medical Research Institute, California
| |
Collapse
|
20
|
Chen L, Liu GQ, Wu HY, Jin J, Yin X, Li D, Lu PR. Monocyte chemoattractant protein 1 and fractalkine play opposite roles in angiogenesis via recruitment of different macrophage subtypes. Int J Ophthalmol 2018; 11:216-222. [PMID: 29487809 DOI: 10.18240/ijo.2018.02.06] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 12/08/2017] [Indexed: 12/13/2022] Open
Abstract
AIM To explore the interaction between macrophages and chemokines [monocyte chemoattractant protein 1 (MCP-1/CCL2) and fractalkine/CX3CL1] and the effects of their interaction on neovascularization. METHODS Human peripheral blood mononuclear cells, donated by healthy volunteers, were separated and cultured in RPMI-1640 medium containing 10% fetal bovine serum, then induced into macrophages by stimulation with 30 µg/L granulocyte macrophage-colony stimulating factor (GM-CSF). The expression of CCR2 and/or CX3CR1 in the macrophages was examined using flow cytometry. Macrophages were then stimulated with recombinant human CCL2 (rh-CCL2) or recombinant human CX3CL1 (rh-CX3CL1). The expression of angiogenesis-related genes, including VEGF-A, THBS-1 and ADAMTS-1 were examined using real-time quantitative polymerase chain reaction (PCR). Supernatants from stimulated macrophages were used in an assay of human retinal endothelial cell (HREC) proliferation. Finally, stimulated macrophages were co-cultured with HREC in a migration assay. RESULTS The expression rate of CCR2 in macrophages stimulated by GM-CSF was 42%±1.9%. The expression rate of CX3CR1 was 71%±3.3%. Compared with vehicle-treated groups, gene expression of VEGF-A in the macrophages was greater in 150 mg/L CCL2-treated groups (P<0.05), while expression of THBS-1 and ADAMTS-1 was significantly lower (P<0.05). By contrast, compared with vehicle-treated groups, expression of VEGF-A in 150 mg/L CX3CL1-treated groups was significantly lower (P<0.05), while expression of THBS-1 and ADAMTS-1 was greater (P<0.05). Supernatants from CCL2 treated macrophages promoted proliferation of HREC (P<0.05), while supernatants from CX3CL1-treated macrophages inhibited the proliferation of HREC (P<0.05). HREC migration increased when co-cultured with CCL2-treated macrophages, but decreased with CX3CL1-treated macrophages (P<0.05). CONCLUSION CCL2 and CX3CL1 exert different effects in regulation of macrophage in expression of angiogenesis-related factors, including VEGF-A, THBS-1 and ADAMTS-1. Our findings suggest that CCL2 and CX3CL1 may be candidate proteins for further exploration of novel targets for treatment of ocular neovascularization.
Collapse
Affiliation(s)
- Lei Chen
- Department of Ophthalmology, the First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Gao-Qin Liu
- Department of Ophthalmology, the First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China.,Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Hong-Ya Wu
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Ji Jin
- Department of Ophthalmology, the First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Xue Yin
- Department of Ophthalmology, the First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Dan Li
- Department of Ophthalmology, the First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Pei-Rong Lu
- Department of Ophthalmology, the First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China.,Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou 215006, Jiangsu Province, China
| |
Collapse
|
21
|
Erythropoietin Signaling Increases Choroidal Macrophages and Cytokine Expression, and Exacerbates Choroidal Neovascularization. Sci Rep 2018; 8:2161. [PMID: 29391474 PMCID: PMC5795007 DOI: 10.1038/s41598-018-20520-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 01/19/2018] [Indexed: 12/14/2022] Open
Abstract
Erythropoietin (EPO) is recognized for neuroprotective and angiogenic effects and has been associated with aging and neovascular age-related macular degeneration (AMD). We hypothesized that systemic EPO facilitates the development of choroidal neovascularization (CNV). Wild type mice expressed murine EPOR (mWtEPOR) in RPE/choroids at baseline and had significantly increased serum EPO after laser treatment. To test the role of EPO signaling, we used human EPOR knock-in mice with the mWtEPOR gene replaced by either the human EPOR gene (hWtEPOR) or a mutated human EPOR gene (hMtEPOR) in a laser-induced choroidal neovascularization (LCNV) model. Loss-of-function hWtEPOR mice have reduced downstream activation, whereas gain-of-function hMtEPOR mice have increased EPOR signaling. Compared to littermate controls (mWtEPOR), hMtEPOR with increased EPOR signaling developed larger CNV lesions. At baseline, hMtEPOR mice had increased numbers of macrophages, greater expression of macrophage markers F4/80 and CD206, and following laser injury, had greater expression of cytokines CCL2, CXCL10, CCL22, IL-6, and IL-10 than mWtEPOR controls. These data support a hypothesis that injury from age- and AMD-related changes in the RPE/choroid leads to choroidal neovascularization through EPOR-mediated cytokine production.
Collapse
|
22
|
On phagocytes and macular degeneration. Prog Retin Eye Res 2017; 61:98-128. [DOI: 10.1016/j.preteyeres.2017.06.002] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 05/29/2017] [Accepted: 06/05/2017] [Indexed: 12/17/2022]
|
23
|
Lechner J, Chen M, Hogg RE, Toth L, Silvestri G, Chakravarthy U, Xu H. Peripheral blood mononuclear cells from neovascular age-related macular degeneration patients produce higher levels of chemokines CCL2 (MCP-1) and CXCL8 (IL-8). J Neuroinflammation 2017; 14:42. [PMID: 28231837 PMCID: PMC5324243 DOI: 10.1186/s12974-017-0820-y] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 02/18/2017] [Indexed: 01/07/2023] Open
Abstract
Background Infiltrating immune cells including monocytes/macrophages have been implicated in the pathogenesis of neovascular age-related macular degeneration (nAMD). The aim of this study was to investigate the cytokine and chemokine expression and secretion profile of peripheral blood mononuclear cells (PBMCs) from nAMD patients and the relationship between the cytokine/chemokine expression profile and clinical phenotype of nAMD, including macular fibrosis, macular atrophy or the responsiveness to anti-VEGF therapy. Methods One hundred sixty-one nAMD patients and 43 controls were enrolled in this study. nAMD patients were divided into subgroups based on the presence/absence of (1) macular atrophy, (2) macular fibrosis and (3) responsiveness to anti-VEGF therapy; 25–30 ml of peripheral blood were obtained from all participants and 5 ml were used for serum collection, and the remaining were used for PBMC isolation using density gradient centrifugation. Intracellular cytokine expressions by PBMCs following phorbol 12-myristate 13-acetate (PMA) and ionomycin stimulation were examined using flow cytometry. Cytokine productions in lipopolysaccharides (LPS)-or 1% oxygen -treated PBMC were measured using cytometric bead array (CBA) assay. In addition, cytokine and chemokine levels in the serum were also measured by CBA assay. Results PBMCs from nAMD patients secreted higher levels of IL-8, CCL2 and VEGF, especially following LPS and 1% oxygen stimulation, than those from controls. 60~80% of IL-8 producing cells were CD11b+CD3− monocytes. The percentage of CD11b+CD3− IL-8+ was significantly increased in nAMD patients compared to controls. PBMCs from nAMD patients without macular fibrosis produced the highest levels of IL-8 and CCL2, whilst PBMCs from nAMD patients with macular atrophy produced highest levels of VEGF. In addition, PBMCs from patients who partially responded to anti-VEGF produced higher levels of IL-8 compared to the cells from complete responders. Interestingly, serum level of CCL2 was not increased in nAMD patients although there was a trend of increased IL-8 in nAMD patients. Conclusions PBMCs, in particular monocytes, may contribute to CNV development in nAMD through secreting elevated levels of IL-8, CCL2 and VEGF after they are recruited to the macula. Apart from VEGF, IL-8 and CCL2 may be additional targets for nAMD management. Electronic supplementary material The online version of this article (doi:10.1186/s12974-017-0820-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Judith Lechner
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - Mei Chen
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - Ruth E Hogg
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - Levente Toth
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - Giuliana Silvestri
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - Usha Chakravarthy
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - Heping Xu
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, UK. .,The Wellcome-Wolfson Institute of Experimental Medicine, Queen's University Belfast, 97 Lisburn Road, BT9 7BL, Belfast, UK.
| |
Collapse
|
24
|
Benhar I, Reemst K, Kalchenko V, Schwartz M. The retinal pigment epithelium as a gateway for monocyte trafficking into the eye. EMBO J 2016; 35:1219-35. [PMID: 27107049 DOI: 10.15252/embj.201694202] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Accepted: 03/21/2016] [Indexed: 11/09/2022] Open
Abstract
The choroid plexus epithelium within the brain ventricles orchestrates blood-derived monocyte entry to the central nervous system under injurious conditions, including when the primary injury site is remote from the brain. Here, we hypothesized that the retinal pigment epithelium (RPE) serves a parallel role, as a gateway for monocyte trafficking to the retina following direct or remote injury. We found elevated expression of genes encoding leukocyte trafficking determinants in mouse RPE as a consequence of retinal glutamate intoxication or optic nerve crush (ONC). Blocking VCAM-1 after ONC interfered with monocyte infiltration into the retina and resulted in a local pro-inflammatory cytokine bias. Live imaging of the injured eye showed monocyte accumulation first in the RPE, and subsequently in the retina, and peripheral leukocytes formed close contact with the RPE Our findings further implied that the ocular milieu can confer monocytes a phenotype advantageous for neuroprotection. These results suggest that the eye utilizes a mechanism of crosstalk with the immune system similar to that of the brain, whereby epithelial barriers serve as gateways for leukocyte entry.
Collapse
Affiliation(s)
- Inbal Benhar
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Kitty Reemst
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Vyacheslav Kalchenko
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | - Michal Schwartz
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|