1
|
Jobling AI, Greferath U, Dixon MA, Quiriconi P, Eyar B, van Koeverden AK, Mills SA, Vessey KA, Bui BV, Fletcher EL. Microglial regulation of the retinal vasculature in health and during the pathology associated with diabetes. Prog Retin Eye Res 2025; 106:101349. [PMID: 40020909 DOI: 10.1016/j.preteyeres.2025.101349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 02/25/2025] [Accepted: 02/25/2025] [Indexed: 03/03/2025]
Abstract
The high metabolic demand of retinal neurons requires a precisely regulated vascular system that can deliver rapid changes in blood flow in response to neural need. In the retina, this is achieved via the action of a coordinated group of cells that form the neurovascular unit. While cells such as pericytes, Müller cells, and astrocytes have long been linked to neurovascular coupling, more recently the resident microglial population have also been implicated. In the healthy retina, microglia make extensive contact with blood vessels, as well as neuronal synapses, and are important in vascular patterning during development. Work in the brain and retina has recently indicated that microglia can directly regulate the local vasculature. In the retina, the fractalkine-Cx3cr1 signalling axis has been shown to induce local capillary constriction within the superficial vascular plexus via a mechanism involving components of the renin-angiotensin system. Furthermore, aberrant microglial induced vasoconstriction may be at the centre of early vascular reactivity changes observed in those with diabetes. This review summarizes the recent emerging evidence that microglia play multiple roles in retinal homeostasis especially in regulating the vasculature. We highlight what is known about the role of microglia under normal circumstances, and then build on this to discuss how microglia contribute to early vascular compromise during diabetes. Further understanding of the mechanisms of microglial-vascular regulation may allow alternate treatment strategies to be devised to reduce vascular pathology in diseases such as diabetic retinopathy.
Collapse
Affiliation(s)
- Andrew I Jobling
- Department of Anatomy and Physiology, The University of Melbourne, Victoria, Australia
| | - Ursula Greferath
- Department of Anatomy and Physiology, The University of Melbourne, Victoria, Australia
| | - Michael A Dixon
- Department of Anatomy and Physiology, The University of Melbourne, Victoria, Australia
| | - Pialuisa Quiriconi
- Department of Anatomy and Physiology, The University of Melbourne, Victoria, Australia
| | - Belinda Eyar
- Department of Anatomy and Physiology, The University of Melbourne, Victoria, Australia
| | - Anna K van Koeverden
- Department of Optometry and Vision Sciences, The University of Melbourne, Victoria, Australia
| | - Samuel A Mills
- Department of Anatomy and Physiology, The University of Melbourne, Victoria, Australia
| | - Kirstan A Vessey
- Department of Anatomy and Physiology, The University of Melbourne, Victoria, Australia
| | - Bang V Bui
- Department of Optometry and Vision Sciences, The University of Melbourne, Victoria, Australia
| | - Erica L Fletcher
- Department of Anatomy and Physiology, The University of Melbourne, Victoria, Australia.
| |
Collapse
|
2
|
Wu M, Fletcher EL, Chinnery HR, Downie LE, Mueller SN. Redefining our vision: an updated guide to the ocular immune system. Nat Rev Immunol 2024; 24:896-911. [PMID: 39215057 DOI: 10.1038/s41577-024-01064-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/01/2024] [Indexed: 09/04/2024]
Abstract
Balanced immune responses in the eyes are crucial to preserve vision. The ocular immune system has long been considered distinct, owing to the so-called 'immune privilege' of its component tissues. More recently, intravital imaging and transcriptomic techniques have reshaped scientific understanding of the ocular immune landscape, such as revealing the specialization of immune cell populations in the various tissues of the eye. As knowledge of the phenotypes of corneal and retinal immune cells has evolved, links to both the systemic immune system, and the central and peripheral nervous systems, have been identified. Using intravital imaging, T cells have recently been found to reside in, and actively patrol, the healthy human cornea. Disease-associated retinal microglia with links to retinal degeneration have also been identified. This Review provides an updated guide to the ocular immune system, highlighting current knowledge of the immune cells that are present in steady-state and specific diseased ocular tissues, as well as evidence for their relationship to systemic disease. In addition, we discuss emerging intravital imaging techniques that can be used to visualize immune cell morphology and dynamics in living human eyes and how these could be applied to advance understanding of the human immune system.
Collapse
Affiliation(s)
- Mengliang Wu
- Department of Optometry and Vision Sciences, The University of Melbourne, Carlton, Victoria, Australia
- Department of Microbiology and Immunology, The University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Erica L Fletcher
- Department of Anatomy and Physiology, The University of Melbourne, Carlton, Victoria, Australia
| | - Holly R Chinnery
- Department of Optometry and Vision Sciences, The University of Melbourne, Carlton, Victoria, Australia.
- Lions Eye Institute, Nedlands, Western Australia, Australia.
- Optometry, The University of Western Australia, Crawley, Western Australia, Australia.
| | - Laura E Downie
- Department of Optometry and Vision Sciences, The University of Melbourne, Carlton, Victoria, Australia.
| | - Scott N Mueller
- Department of Microbiology and Immunology, The University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia.
| |
Collapse
|
3
|
Eugenín J, Eugenín-von Bernhardi L, von Bernhardi R. Age-dependent changes on fractalkine forms and their contribution to neurodegenerative diseases. Front Mol Neurosci 2023; 16:1249320. [PMID: 37818457 PMCID: PMC10561274 DOI: 10.3389/fnmol.2023.1249320] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 09/06/2023] [Indexed: 10/12/2023] Open
Abstract
The chemokine fractalkine (FKN, CX3CL1), a member of the CX3C subfamily, contributes to neuron-glia interaction and the regulation of microglial cell activation. Fractalkine is expressed by neurons as a membrane-bound protein (mCX3CL1) that can be cleaved by extracellular proteases generating several sCX3CL1 forms. sCX3CL1, containing the chemokine domain, and mCX3CL1 have high affinity by their unique receptor (CX3CR1) which, physiologically, is only found in microglia, a resident immune cell of the CNS. The activation of CX3CR1contributes to survival and maturation of the neural network during development, glutamatergic synaptic transmission, synaptic plasticity, cognition, neuropathic pain, and inflammatory regulation in the adult brain. Indeed, the various CX3CL1 forms appear in some cases to serve an anti-inflammatory role of microglia, whereas in others, they have a pro-inflammatory role, aggravating neurological disorders. In the last decade, evidence points to the fact that sCX3CL1 and mCX3CL1 exhibit selective and differential effects on their targets. Thus, the balance in their level and activity will impact on neuron-microglia interaction. This review is focused on the description of factors determining the emergence of distinct fractalkine forms, their age-dependent changes, and how they contribute to neuroinflammation and neurodegenerative diseases. Changes in the balance among various fractalkine forms may be one of the mechanisms on which converge aging, chronic CNS inflammation, and neurodegeneration.
Collapse
Affiliation(s)
- Jaime Eugenín
- Facultad de Química y Biología, Departamento de Biología, Universidad de Santiago de Chile, USACH, Santiago, Chile
| | | | - Rommy von Bernhardi
- Facultad de Ciencias para el Cuidado de la Salud, Universidad San Sebastián, Santiago, Chile
| |
Collapse
|
4
|
Jong ED, Hacibekiroglu S, Guo L, Sawula E, Li B, Li C, Ho MT, Shoichet MS, Wallace VA, Nagy A. Soluble CX3CL1-expressing retinal pigment epithelium cells protect rod photoreceptors in a mouse model of retinitis pigmentosa. Stem Cell Res Ther 2023; 14:212. [PMID: 37605279 PMCID: PMC10441732 DOI: 10.1186/s13287-023-03434-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 07/26/2023] [Indexed: 08/23/2023] Open
Abstract
BACKGROUND Retinitis pigmentosa (RP) is an inherited retinal disease that results in photoreceptor degeneration, leading to severe vision loss or blindness. Due to its genetic heterogeneity, developing a new gene therapy to correct every genetic mutation contributing to its progression is infeasible. Photoreceptor transplantation can be harnessed to restore vision; however, this approach is limited by poor cell survival and synaptic integration into the neural retina. Thus, we developed a combined cell and gene therapy that is expected to protect photoreceptors in most, if not all, cases of RP. METHODS Human embryonic stem cells (hESCs) modified with our FailSafe™ system were genetically engineered to overexpress sCX3CL1, an inhibitor of microglia activation that has been shown to preserve photoreceptor survival and function in mouse models of RP, independent of the genetic cause. These cells were differentiated into human retinal pigment epithelium (hRPE) cells and used as therapeutic cells due to their longevity and safety, both of which have been demonstrated in preclinical and clinical studies. Transgenic hRPE were delivered into the subretinal space of immunodeficient mice and the rd10 mouse model of RP to evaluate donor cell survival and retention of transgene expression. The outer nuclear layer was quantified to assess photoreceptor protection. RESULTS Transgenic FailSafe™ hRPE (FS-hRPE) cells can survive for at least four months in the retina of immunodeficient mice and retain transgene expression. However, these cells do not persist beyond two weeks post-injection in the retina of immunocompetent rd10 recipients, despite Cyclosporine A treatment. Nevertheless, sCX3CL1-expressing FailSafe™ hRPE cells prevented photoreceptor degeneration in a local acting manner during the duration of their presence in the subretinal space. CONCLUSIONS Transgenic hESCs differentiate into hRPE cells and retain sCX3CL1 transgene expression both in vitro and in vivo. Moreover, hRPE cells delivered to the subretinal space of rd10 mice prevented photoreceptor degeneration in a local-acting manner, suggesting that this approach could have applications for preserving photoreceptors in specific subregions of the retina, such as the macula. Overall, our study not only reveals the potential of a combined cell and gene therapy for the treatment of RP, but also the possibility of using hRPE cells to deliver therapeutic biologics in situ to treat diseases over long-term.
Collapse
Affiliation(s)
- Eric D Jong
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto25 Orde St, 5Th Floor, Room 5-1015, Toronto, ON, M5T 3H7, Canada
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Sabiha Hacibekiroglu
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto25 Orde St, 5Th Floor, Room 5-1015, Toronto, ON, M5T 3H7, Canada
| | - Lily Guo
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto25 Orde St, 5Th Floor, Room 5-1015, Toronto, ON, M5T 3H7, Canada
| | - Evan Sawula
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto25 Orde St, 5Th Floor, Room 5-1015, Toronto, ON, M5T 3H7, Canada
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Biao Li
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto25 Orde St, 5Th Floor, Room 5-1015, Toronto, ON, M5T 3H7, Canada
| | - Chengjin Li
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto25 Orde St, 5Th Floor, Room 5-1015, Toronto, ON, M5T 3H7, Canada
| | - Margaret T Ho
- Institute of Biomedical Engineering, University of Toronto, Toronto, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Canada
| | - Molly S Shoichet
- Institute of Biomedical Engineering, University of Toronto, Toronto, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Canada
- Department of Chemistry, University of Toronto, Toronto, Canada
| | - Valerie A Wallace
- Donald K. Johnson Eye Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Department of Ophthalmology and Vision Sciences, University of Toronto, Toronto, ON, Canada
| | - Andras Nagy
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto25 Orde St, 5Th Floor, Room 5-1015, Toronto, ON, M5T 3H7, Canada.
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
- Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia.
- Department of Obstetrics & Gynecology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
5
|
Qi Y, Liu L, Liang D, Tang S, Yu X, Ye H, Chen N. Bujing Yishi tablets alleviate photoreceptor cells death via the P2X7R/CX3CL1/CX3CR1 pathway in Retinitis Pigmentosa rats. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 115:154828. [PMID: 37116386 DOI: 10.1016/j.phymed.2023.154828] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 04/07/2023] [Accepted: 04/16/2023] [Indexed: 05/21/2023]
Abstract
BACKGROUND Retinitis pigmentosa (RP) refers to a group of progressive photoreceptor degenerative diseases. The activation of microglia has been reported to play an important role in the photoreceptor degeneration in RP retinal. Bujing Yishi tablets (BJYS), a Chinese herbal medicine, has been used to treat retinal diseases in China with desirable effect in improving visual function. However, the mechanisms underlying the efficacy of BJYS treatment in RP are not yet fully understood. PURPOSE Based on the preliminary experiments, this study aimed to investigate the therapeutic mechanism involved in treating N-Methyl-N-Nitrosourea (MNU)-induced retinal degeneration of RP with BJYS. METHODS To explore the efficacy of BJYS, a rat experimental RP model was established through intraperitoneal injection of MNU (50 mg/kg). Two experiment was carried out. After the treatment, we conducted H&E, TUNEL, retinal cytokine levels and IBA-1 expression in microglia to confirm the impact on RP model. The specific mechanism of action of BJYS tablet was assessed by western blot, real-time polymerase chain reaction (RT-PCR), and immunofluorescence to determine the mRNA and protein expression levels involved in clarifying the effectiveness of BJYS exerted through P2X7R/CX3CL1/CX3CR1 pathway. RESULTS Significant alleviation of retinal morphological structure and photoreceptor degeneration by BJYS treatment was observed in the retinal of MNU-induced RP rats, BJYS prevented the reduction of ONL thickness and decreased the level of apoptotic cells in ONL. It also inhibited microglia overactivation and reduced retinal levels of IL-1β, IL-6, TNF-α. In addition, BJYS decreased the protein expression and mRNA expression of P2X7, CX3CL1 and CX3CR1 and reduced the phosphorylation of p38 MAPK. CONCLUSION In summary, this study suggested that BJYS treatment could alleviate photoreceptors degeneration of RP by inhibiting microglia overactivation and inflammation through the P2X7R/CX3CL1/CX3CR1 pathway. These effects suggest that BJYS tablets may serve as a promising oral therapeutic agent for RP.
Collapse
Affiliation(s)
- Yulin Qi
- Department of Ophthalmology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China; Postdoctoral Research Station of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lu Liu
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Dan Liang
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shiyun Tang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaoyi Yu
- Department of Ophthalmology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Hejiang Ye
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China..
| | - Nianzhi Chen
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
6
|
Padovani-Claudio DA, Ramos CJ, Capozzi ME, Penn JS. Elucidating glial responses to products of diabetes-associated systemic dyshomeostasis. Prog Retin Eye Res 2023; 94:101151. [PMID: 37028118 PMCID: PMC10683564 DOI: 10.1016/j.preteyeres.2022.101151] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 04/08/2023]
Abstract
Diabetic retinopathy (DR) is a leading cause of blindness in working age adults. DR has non-proliferative stages, characterized in part by retinal neuroinflammation and ischemia, and proliferative stages, characterized by retinal angiogenesis. Several systemic factors, including poor glycemic control, hypertension, and hyperlipidemia, increase the risk of DR progression to vision-threatening stages. Identification of cellular or molecular targets in early DR events could allow more prompt interventions pre-empting DR progression to vision-threatening stages. Glia mediate homeostasis and repair. They contribute to immune surveillance and defense, cytokine and growth factor production and secretion, ion and neurotransmitter balance, neuroprotection, and, potentially, regeneration. Therefore, it is likely that glia orchestrate events throughout the development and progression of retinopathy. Understanding glial responses to products of diabetes-associated systemic dyshomeostasis may reveal novel insights into the pathophysiology of DR and guide the development of novel therapies for this potentially blinding condition. In this article, first, we review normal glial functions and their putative roles in the development of DR. We then describe glial transcriptome alterations in response to systemic circulating factors that are upregulated in patients with diabetes and diabetes-related comorbidities; namely glucose in hyperglycemia, angiotensin II in hypertension, and the free fatty acid palmitic acid in hyperlipidemia. Finally, we discuss potential benefits and challenges associated with studying glia as targets of DR therapeutic interventions. In vitro stimulation of glia with glucose, angiotensin II and palmitic acid suggests that: 1) astrocytes may be more responsive than other glia to these products of systemic dyshomeostasis; 2) the effects of hyperglycemia on glia are likely to be largely osmotic; 3) fatty acid accumulation may compound DR pathophysiology by promoting predominantly proinflammatory and proangiogenic transcriptional alterations of macro and microglia; and 4) cell-targeted therapies may offer safer and more effective avenues for DR treatment as they may circumvent the complication of pleiotropism in retinal cell responses. Although several molecules previously implicated in DR pathophysiology are validated in this review, some less explored molecules emerge as potential therapeutic targets. Whereas much is known regarding glial cell activation, future studies characterizing the role of glia in DR and how their activation is regulated and sustained (independently or as part of retinal cell networks) may help elucidate mechanisms of DR pathogenesis and identify novel drug targets for this blinding disease.
Collapse
Affiliation(s)
- Dolly Ann Padovani-Claudio
- Department of Ophthalmology and Visual Sciences, Vanderbilt University School of Medicine, B3321A Medical Center North, 1161 21st Avenue South, Nashville, TN, 37232-0011, USA.
| | - Carla J Ramos
- Department of Ophthalmology and Visual Sciences, Vanderbilt University School of Medicine, AA1324 Medical Center North, 1161 21st Avenue South, Nashville, TN, 37232-0011, USA.
| | - Megan E Capozzi
- Duke Molecular Physiology Institute, Duke University School of Medicine, 300 North Duke Street, Durham, NC, 27701, USA.
| | - John S Penn
- Department of Ophthalmology and Visual Sciences, Vanderbilt University School of Medicine, B3307 Medical Center North, 1161 21st Avenue South, Nashville, TN, 37232-0011, USA.
| |
Collapse
|
7
|
Zhao L, Hou C, Yan N. Neuroinflammation in retinitis pigmentosa: Therapies targeting the innate immune system. Front Immunol 2022; 13:1059947. [PMID: 36389729 PMCID: PMC9647059 DOI: 10.3389/fimmu.2022.1059947] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 10/17/2022] [Indexed: 11/13/2022] Open
Abstract
Retinitis pigmentosa (RP) is an important cause of irreversible blindness worldwide and lacks effective treatment strategies. Although mutations are the primary cause of RP, research over the past decades has shown that neuroinflammation is an important cause of RP progression. Due to the abnormal activation of immunity, continuous sterile inflammation results in neuron loss and structural destruction. Therapies targeting inflammation have shown their potential to attenuate photoreceptor degeneration in preclinical models. Regardless of variations in genetic background, inflammatory modulation is emerging as an important role in the treatment of RP. We summarize the evidence for the role of inflammation in RP and mention therapeutic strategies where available, focusing on the modulation of innate immune signals, including TNFα signaling, TLR signaling, NLRP3 inflammasome activation, chemokine signaling and JAK/STAT signaling. In addition, we describe epigenetic regulation, the gut microbiome and herbal agents as prospective treatment strategies for RP in recent advances.
Collapse
Affiliation(s)
- Ling Zhao
- Research Laboratory of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China,Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| | - Chen Hou
- Research Laboratory of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China,Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| | - Naihong Yan
- Research Laboratory of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China,*Correspondence: Naihong Yan,
| |
Collapse
|
8
|
Agarwal P, Agarwal R. Tackling retinal ganglion cell apoptosis in glaucoma: role of adenosine receptors. Expert Opin Ther Targets 2021; 25:585-596. [PMID: 34402357 DOI: 10.1080/14728222.2021.1969362] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION The role of adenosine receptors as therapeutic targets for neuroprotection is now widely recognized. Their role, however, in protection against retinal ganglion cell (RGC) apoptosis in glaucoma needs further investigation. Hence, in this review, we look into the possibility of adenosine receptors as potential therapeutic targets by exploring their role in modulating various pathophysiological mechanisms underlying glaucomatous RGC loss. AREAS COVERED This review presents a summary of the adenosine receptor distribution in retina and the cellular functions mediated by them. The major pathophysiological mechanisms such as excitotoxicity, vascular dysregulation, loss of neurotrophic signaling, and inflammatory responses involved in glaucomatous RGC loss are discussed. The literature showing the role of adenosine receptors in modulating these pathophysiological mechanisms is discussed. The literature search was conducted using Pubmed search engine using key words such as 'RGC apoptosis,' 'adenosine,' adenosine receptors' 'retina' 'excitotoxicity,' 'neurotrophins,' 'ischemia', and 'cytokines' individually and in various combinations. EXPERT OPINION Use of adenosine receptor agonists and antagonists, for preservation of the RGCs in glaucomatous eyes independent of the level of intraocular pressure seems a very useful strategy. Future application of this strategy would require appropriate designing of drug formulation for tissue and disease-specific receptor targeting. Furthermore, the modulation of physiological functions and potential adverse effects need further investigations.
Collapse
Affiliation(s)
- Puneet Agarwal
- School of Medicine, International Medical University, Kuala Lumpur, Malaysia
| | - Renu Agarwal
- School of Medicine, International Medical University, Kuala Lumpur, Malaysia
| |
Collapse
|
9
|
Chamera K, Szuster-Głuszczak M, Basta-Kaim A. Shedding light on the role of CX3CR1 in the pathogenesis of schizophrenia. Pharmacol Rep 2021; 73:1063-1078. [PMID: 34021899 PMCID: PMC8413165 DOI: 10.1007/s43440-021-00269-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 04/22/2021] [Accepted: 04/26/2021] [Indexed: 11/24/2022]
Abstract
Schizophrenia has a complex and heterogeneous molecular and clinical picture. Over the years of research on this disease, many factors have been suggested to contribute to its pathogenesis. Recently, the inflammatory processes have gained particular interest in the context of schizophrenia due to the increasing evidence from epidemiological, clinical and experimental studies. Within the immunological component, special attention has been brought to chemokines and their receptors. Among them, CX3C chemokine receptor 1 (CX3CR1), which belongs to the family of seven-transmembrane G protein-coupled receptors, and its cognate ligand (CX3CL1) constitute a unique system in the central nervous system. In the view of regulation of the brain homeostasis through immune response, as well as control of microglia reactivity, the CX3CL1–CX3CR1 system may represent an attractive target for further research and schizophrenia treatment. In the review, we described the general characteristics of the CX3CL1–CX3CR1 axis and the involvement of this signaling pathway in the physiological processes whose disruptions are reported to participate in mechanisms underlying schizophrenia. Furthermore, based on the available clinical and experimental data, we presented a guide to understanding the implication of the CX3CL1–CX3CR1 dysfunctions in the course of schizophrenia.
Collapse
Affiliation(s)
- Katarzyna Chamera
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St., 31-343, Kraków, Poland.
| | - Magdalena Szuster-Głuszczak
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St., 31-343, Kraków, Poland
| | - Agnieszka Basta-Kaim
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St., 31-343, Kraków, Poland
| |
Collapse
|
10
|
Makabe K, Sugita S, Mandai M, Futatsugi Y, Takahashi M. Microglia dynamics in retinitis pigmentosa model: formation of fundus whitening and autofluorescence as an indicator of activity of retinal degeneration. Sci Rep 2020; 10:14700. [PMID: 32895435 PMCID: PMC7477572 DOI: 10.1038/s41598-020-71626-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Accepted: 06/04/2020] [Indexed: 01/04/2023] Open
Abstract
In patients with retinitis pigmentosa (RP), color fundus photography and fundus autofluorescence (FAF) have been used to estimate the disease progression. To understand the origin and the diagnostic interpretation of the fundus color and FAF, we performed in vivo imaging of fundus color and FAF together with histological analyses of the retinal degeneration process using the RP model mice, rd10. FAF partly represented the accumulation of microglia in the photoreceptor outer segments. Fundus whitening suggested the presence of apoptotic cells, which spatiotemporally preceded increase in FAF. We observed two patterns of FAF localization, arcuate and diffuse, each indicating different pattern of apoptosis, wavy and diffuse, respectively. Diffuse pattern of apoptosis was suppressed in dark-raised rd10 mice, in which outer nuclear layer (ONL) loss was significantly suppressed. The occupancy of FAF correlated with the thinning rate of the ONL. Fractalkine, a microglia chemotactic factor, was detected in apoptotic photoreceptors, suggesting chemokine-induced recruitment of microglia into the ONL, which paralleled with accelerated ONL loss and increased FAF occupancy. Thus, we propose that the degree of photoreceptor apoptosis and the rate of ONL thinning in RP patients might be read from the fundus color and the FAF.
Collapse
Affiliation(s)
- Kenichi Makabe
- Laboratory for Retinal Regeneration, Center for Biosystems Dynamics Research, RIKEN, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan.,Department of Ophthalmology, Kobe Kaisei Hospital, 3-11-15 Shinoharakitamachi, Nada-ku, Kobe, 657-0068, Japan
| | - Sunao Sugita
- Laboratory for Retinal Regeneration, Center for Biosystems Dynamics Research, RIKEN, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan.
| | - Michiko Mandai
- Laboratory for Retinal Regeneration, Center for Biosystems Dynamics Research, RIKEN, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan
| | - Yoko Futatsugi
- Laboratory for Retinal Regeneration, Center for Biosystems Dynamics Research, RIKEN, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan
| | - Masayo Takahashi
- Laboratory for Retinal Regeneration, Center for Biosystems Dynamics Research, RIKEN, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan
| |
Collapse
|
11
|
Lavalette S, Conart JB, Touhami S, Roubeix C, Houssier M, Augustin S, Raoul W, Combadière C, Febbraio M, Ong H, Chemtob S, Sahel JA, Delarasse C, Guillonneau X, Sennlaub F. CD36 Deficiency Inhibits Retinal Inflammation and Retinal Degeneration in Cx3cr1 Knockout Mice. Front Immunol 2020; 10:3032. [PMID: 31969887 PMCID: PMC6960398 DOI: 10.3389/fimmu.2019.03032] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 12/10/2019] [Indexed: 12/22/2022] Open
Abstract
Background: CD36, a member of the class B scavenger receptor family, participates in Toll-like receptor signaling on mononuclear phagocytes (MP) and can promote sterile pathogenic inflammation. We here analyzed the effect of CD36 deficiency on retinal inflammation and photoreceptor degeneration, the hallmarks of age-related macular degeneration (AMD), that characterize Cx3cr1−/−mice. Methods: We analyzed subretinal MP accumulation, and cone- and rod-degeneration in light-challenged and aged, CD36 competent or deficient, hyper-inflammatory Cx3cr1−/− mice, using histology and immune-stained retinal flatmounts. Monocytes (Mo) were subretinally adoptively transferred to evaluate their elimination rate from the subretinal space and Interleukin 6 (IL-6) secretion from cultured Mo-derived cells (MdCs) of the different mouse strains were analyzed. Results: CD36 deficient Cx3cr1−/− mice were protected against age- and light-induced subretinal inflammation and associated cone and rod degeneration. CD36 deficiency in Cx3cr1−/− MPs inhibited their prolonged survival in the immune-suppressive subretinal space and reduced the exaggerated IL-6 secretion observed in Cx3cr1−/− MPs that we previously showed leads to increased subretinal MP survival. Conclusion:Cd36 deficiency significantly protected hyperinflammatory Cx3cr1−/− mice against subretinal MP accumulation and associated photoreceptor degeneration. The observed CD36-dependent induction of pro-inflammatory IL-6 might be at least partially responsible for the prolonged MP survival in the immune-suppressive environment and its pathological consequences on photoreceptor homeostasis.
Collapse
Affiliation(s)
- Sophie Lavalette
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France
| | | | - Sara Touhami
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France
| | | | - Marianne Houssier
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France
| | | | - William Raoul
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France.,Université de Tours, Inserm, N2C UMR 1069, Faculté de Médecine, Tours, France
| | - Christophe Combadière
- Sorbonne Université, Inserm, CNRS, Centre d'Immunologie et des Maladies Infectieuses, Cimi-Paris, Paris, France
| | - Maria Febbraio
- Department of Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Huy Ong
- Faculty of Pharmacy, Université de Montréal, Montreal, QC, Canada
| | - Sylvain Chemtob
- Departments of Pediatrics, Ophthalmology and Pharmacology, Université de Montréal, Montreal, QC, Canada
| | - José-Alain Sahel
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France
| | - Cécile Delarasse
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France
| | | | - Florian Sennlaub
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France
| |
Collapse
|
12
|
Chamera K, Trojan E, Szuster-Głuszczak M, Basta-Kaim A. The Potential Role of Dysfunctions in Neuron-Microglia Communication in the Pathogenesis of Brain Disorders. Curr Neuropharmacol 2020; 18:408-430. [PMID: 31729301 PMCID: PMC7457436 DOI: 10.2174/1570159x17666191113101629] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 10/15/2019] [Accepted: 11/10/2019] [Indexed: 12/18/2022] Open
Abstract
The bidirectional communication between neurons and microglia is fundamental for the proper functioning of the central nervous system (CNS). Chemokines and clusters of differentiation (CD) along with their receptors represent ligand-receptor signalling that is uniquely important for neuron - microglia communication. Among these molecules, CX3CL1 (fractalkine) and CD200 (OX-2 membrane glycoprotein) come to the fore because of their cell-type-specific localization. They are principally expressed by neurons when their receptors, CX3CR1 and CD200R, respectively, are predominantly present on the microglia, resulting in the specific axis which maintains the CNS homeostasis. Disruptions to this balance are suggested as contributors or even the basis for many neurological diseases. In this review, we discuss the roles of CX3CL1, CD200 and their receptors in both physiological and pathological processes within the CNS. We want to underline the critical involvement of these molecules in controlling neuron - microglia communication, noting that dysfunctions in their interactions constitute a key factor in severe neurological diseases, such as schizophrenia, depression and neurodegeneration-based conditions.
Collapse
Affiliation(s)
- Katarzyna Chamera
- Department of Experimental Neuroendocrinology, Laboratory of Immunoendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St. 31-343Kraków, Poland
| | - Ewa Trojan
- Department of Experimental Neuroendocrinology, Laboratory of Immunoendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St. 31-343Kraków, Poland
| | - Magdalena Szuster-Głuszczak
- Department of Experimental Neuroendocrinology, Laboratory of Immunoendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St. 31-343Kraków, Poland
| | - Agnieszka Basta-Kaim
- Department of Experimental Neuroendocrinology, Laboratory of Immunoendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St. 31-343Kraków, Poland
| |
Collapse
|
13
|
Rashid K, Akhtar-Schaefer I, Langmann T. Microglia in Retinal Degeneration. Front Immunol 2019; 10:1975. [PMID: 31481963 PMCID: PMC6710350 DOI: 10.3389/fimmu.2019.01975] [Citation(s) in RCA: 233] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 08/05/2019] [Indexed: 12/18/2022] Open
Abstract
The retina is a complex tissue with multiple cell layers that are highly ordered. Its sophisticated structure makes it especially sensitive to external or internal perturbations that exceed the homeostatic range. This necessitates the continuous surveillance of the retina for the detection of noxious stimuli. This task is mainly performed by microglia cells, the resident tissue macrophages which confer neuroprotection against transient pathophysiological insults. However, under sustained pathological stimuli, microglial inflammatory responses become dysregulated, often worsening disease pathology. In this review, we provide an overview of recent studies that depict microglial responses in diverse retinal pathologies that have degeneration and chronic immune reactions as key pathophysiological components. We also discuss innovative immunomodulatory therapy strategies that dampen the detrimental immunological responses to improve disease outcome.
Collapse
Affiliation(s)
- Khalid Rashid
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Isha Akhtar-Schaefer
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Thomas Langmann
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, Cologne, Germany
| |
Collapse
|
14
|
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: 95] [Impact Index Per Article: 15.8] [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
|
15
|
Liu JQ, Chu SF, Zhou X, Zhang DY, Chen NH. Role of chemokines in Parkinson's disease. Brain Res Bull 2019; 152:11-18. [PMID: 31136787 DOI: 10.1016/j.brainresbull.2019.05.020] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Revised: 05/17/2019] [Accepted: 05/23/2019] [Indexed: 01/10/2023]
Abstract
Parkinson's disease (PD) is a chronic progressive neurodegenerative disorder with an increasing incidence year by year, particularly as the population ages. The most common neuropathologic manifestation in patients with Parkinson's disease is dopamine neurons degeneration and loss in substantia nigra of middle brain. The main neurochemistry problem is the lack of the neurotransmitter dopamine. Clinically, PD patients may also have higher levels of glutamate, gamma-aminobutyric acid, acetylcholine and other neurotransmitters. At present, many data have shown that some chemokines are involved in regulating the release and transmission of neurotransmitters, and the growth and development of related neurons. In recent years, most of the studies relative to PD is based on immune and inflammatory mechanisms, and chemokines is also the focus on this mechanism. Chemokines are a class of cytokines that have definite chemotaxis effects on the different target cells. They might be involved in the pathogenesis of PD by inducing neuronal apoptosis and microglia activation. Clinical data has shown that the levels of chemokines in plasma and cerebrospinal fluid of PD patients have corresponding changes compared with the healthy persons. This review summarizes recent studies on chemokines and their receptors in Parkinson's disease: (i) to explore the role of chemokines in Parkinson's disease; (ii) to provide new indicators for clinical diagnosis of PD; (iii) to provide new targets for drug research and development in the treatment of Parkinson's disease.
Collapse
Affiliation(s)
- Jia-Qi Liu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; Institute of Pharmaceutical Science, China Pharmaceutical University, Nanjing 211198, China
| | - Shi-Feng Chu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Xin Zhou
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Da-Yong Zhang
- Institute of Pharmaceutical Science, China Pharmaceutical University, Nanjing 211198, China
| | - Nai-Hong Chen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
16
|
Central fractalkine stimulates central prostaglandin E 2 production and induces systemic inflammatory responses. Brain Res Bull 2018; 140:311-317. [PMID: 29870777 DOI: 10.1016/j.brainresbull.2018.05.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 04/23/2018] [Accepted: 05/31/2018] [Indexed: 11/20/2022]
Abstract
Fractalkine (FKN; CX3CL1) belongs to gamma-chemokine family and binds to CX3CR1 receptors. Currently, the mechanisms involving FKN-induced inflammatory mediators are research targets in an attempt to study immune diseases mechanisms. Besides, FKN seems to modulate inflammation in the nervous system by inducing the secretion of pro-inflammatory mediators such as prostaglandin E2 (PGE2). PGE2 is a classic and important mediator of fever that activates warm-responsive neurons in the anteroventral preoptic region of the hypothalamus (AVPO). Here, we tested the hypothesis that central FKN modulates febrigenic signaling both centrally and peripherally. We performed intracerebroventricular (icv) microinjections of saline (1 μL) or FKN (doses of 5, 50, 500 pg/μL) in rats and measured body temperature (Tb) besides assessing tail skin temperature (Tsk) as a thermoeffector indicator used to calculate the heat loss index (HLI). We also measured the time course changes in AVPO PGE2, besides plasma corticosterone (CORT) and interleukin-6 (IL-6) levels. FKN induced a long lasting febrile response in which the highest dose (500 pg/μL) induced a marked rise on Tb that was accompanied by a reduced Tsk and HLI, consequently. FKN increased AVPO PGE2 production in a time-dependent manner besides increasing plasma CORT and IL-6 levels. Our data consistently indicate that FKN increases AVPO PGE2 production and Tb, accompanied by raised plasma IL-6 levels and activation of the hypothalamus-pituitary-adrenal axis.
Collapse
|
17
|
The Role of the Microglial Cx3cr1 Pathway in the Postnatal Maturation of Retinal Photoreceptors. J Neurosci 2018; 38:4708-4723. [PMID: 29669747 DOI: 10.1523/jneurosci.2368-17.2018] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 03/22/2018] [Accepted: 04/10/2018] [Indexed: 11/21/2022] Open
Abstract
Microglia are the resident immune cells of the CNS, and their response to infection, injury and disease is well documented. More recently, microglia have been shown to play a role in normal CNS development, with the fractalkine-Cx3cr1 signaling pathway of particular importance. This work describes the interaction between the light-sensitive photoreceptors and microglia during eye opening, a time of postnatal photoreceptor maturation. Genetic removal of Cx3cr1 (Cx3cr1GFP/GFP ) led to an early retinal dysfunction soon after eye opening [postnatal day 17 (P17)] and cone photoreceptor loss (P30 onward) in mice of either sex. This dysfunction occurred at a time when fractalkine expression was predominantly outer retinal, when there was an increased microglial presence near the photoreceptor layer and increased microglial-cone photoreceptor contacts. Photoreceptor maturation and outer segment elongation was coincident with increased opsin photopigment expression in wild-type retina, while this was aberrant in the Cx3cr1GFP/GFP retina and outer segment length was reduced. A beadchip array highlighted Cx3cr1 regulation of genes involved in the photoreceptor cilium, a key structure that is important for outer segment elongation. This was confirmed with quantitative PCR with specific cilium-related genes, Rpgr and Rpgrip1, downregulated at eye opening (P14). While the overall cilium structure was unaffected, expression of Rpgr, Rpgrip1, and centrin were restricted to more proximal regions of the transitional zone. This study highlighted a novel role for microglia in postnatal neuronal development within the retina, with loss of fractalkine-Cx3cr1 signaling leading to an altered distribution of cilium proteins, failure of outer segment elongation and ultimately cone photoreceptor loss.SIGNIFICANCE STATEMENT Microglia are involved in CNS development and disease. This work highlights the role of microglia in postnatal development of the light-detecting photoreceptor neurons within the mouse retina. Loss of the microglial Cx3cr1 signaling pathway resulted in specific alterations in the cilium, a key structure in photoreceptor outer segment elongation. The distribution of key components of the cilium transitional zone, Rpgr, Rpgrip1, and centrin, were altered in retinae lacking Cx3cr1 with reduced outer segment length and cone photoreceptor death observed at later postnatal ages. This work identifies a novel role for microglia in the postnatal maturation of retinal photoreceptors.
Collapse
|
18
|
Zhang Y, Zhao L, Wang X, Ma W, Lazere A, Qian HH, Zhang J, Abu-Asab M, Fariss RN, Roger JE, Wong WT. Repopulating retinal microglia restore endogenous organization and function under CX3CL1-CX3CR1 regulation. SCIENCE ADVANCES 2018; 4:eaap8492. [PMID: 29750189 PMCID: PMC5943055 DOI: 10.1126/sciadv.aap8492] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 02/12/2018] [Indexed: 05/30/2023]
Abstract
Microglia have been discovered to undergo repopulation following ablation. However, the functionality of repopulated microglia and the mechanisms regulating microglia repopulation are unknown. We examined microglial homeostasis in the adult mouse retina, a specialized neural compartment containing regular arrays of microglia in discrete synaptic laminae that can be directly visualized. Using in vivo imaging and cell-fate mapping techniques, we discovered that repopulation originated from residual microglia proliferating in the central inner retina that subsequently spread by centrifugal migration to fully recapitulate pre-existing microglial distributions and morphologies. Repopulating cells fully restored microglial functions including constitutive "surveying" process movements, behavioral and physiological responses to retinal injury, and maintenance of synaptic structure and function. Microglial repopulation was regulated by CX3CL1-CX3CR1 signaling, slowing in CX3CR1 deficiency and accelerating with exogenous CX3CL1 administration. Microglial homeostasis following perturbation can fully recover microglial organization and function under the regulation of chemokine signaling between neurons and microglia.
Collapse
Affiliation(s)
- Yikui Zhang
- Unit on Neuron-Glia Interactions in Retinal Disease, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
- The Eye Hospital, School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou 325027, China
| | - Lian Zhao
- Unit on Neuron-Glia Interactions in Retinal Disease, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Xu Wang
- Unit on Neuron-Glia Interactions in Retinal Disease, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Wenxin Ma
- Unit on Neuron-Glia Interactions in Retinal Disease, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Adam Lazere
- Unit on Neuron-Glia Interactions in Retinal Disease, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Hao-hua Qian
- Visual Function Core, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jun Zhang
- Synaptic Physiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mones Abu-Asab
- Section on Histopathology, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Robert N. Fariss
- Biological Imaging Core, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jerome E. Roger
- Centre d’Etude et de Recherche Thérapeutique en Ophtalmologie (CERTO), Retina France, Orsay, France
- Paris-Saclay Institute of Neuroscience, CNRS, Université Paris-Sud, Université Paris-Saclay, Orsay, France
| | - Wai T. Wong
- Unit on Neuron-Glia Interactions in Retinal Disease, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
19
|
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]
|
20
|
Fractalkine-CX3CR1 signaling is critical for progesterone-mediated neuroprotection in the retina. Sci Rep 2017; 7:43067. [PMID: 28216676 PMCID: PMC5316933 DOI: 10.1038/srep43067] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 01/18/2017] [Indexed: 12/16/2022] Open
Abstract
Retinitis pigmentosa (RP) encompasses a group of retinal diseases resulting in photoreceptor loss and blindness. We have previously shown in the rd10 mouse model of RP, that rd10 microglia drive degeneration of viable neurons. Norgestrel, a progesterone analogue, primes viable neurons against potential microglial damage. In the current study we wished to investigate this neuroprotective effect further. We were particularly interested in the role of fractalkine-CX3CR1 signaling, previously shown to mediate photoreceptor-microglia crosstalk and promote survival in the rd10 retina. Norgestrel upregulates fractalkine-CX3CR1 signaling in the rd10 retina, coinciding with photoreceptor survival. We show that Norgestrel-treated photoreceptor-like cells, 661Ws, and C57 explants modulate rd10 microglial activity in co-culture, resulting in increased photoreceptor survival. Assessment of Norgestrel's neuroprotective effects when fractalkine was knocked-down in 661 W cells and release of fractalkine was reduced in rd10 explants confirms a crucial role for fractalkine-CX3CR1 signaling in Norgestrel-mediated neuroprotection. To further understand the role of fractalkine in neuroprotection, we assessed the release of 40 cytokines in fractalkine-treated rd10 microglia and explants. In both cases, treatment with fractalkine reduced a variety of pro-inflammatory cytokines. These findings further our understanding of Norgestrel's neuroprotective properties, capable of modulating harmful microglial activity indirectly through photoreceptors, leading to increased neuroprotection.
Collapse
|
21
|
Breen KT, Anderson SR, Steele MR, Calkins DJ, Bosco A, Vetter ML. Loss of Fractalkine Signaling Exacerbates Axon Transport Dysfunction in a Chronic Model of Glaucoma. Front Neurosci 2016; 10:526. [PMID: 27932942 PMCID: PMC5123443 DOI: 10.3389/fnins.2016.00526] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Accepted: 10/31/2016] [Indexed: 01/01/2023] Open
Abstract
Neurodegeneration in glaucoma results in decline and loss of retinal ganglion cells (RGCs), and is associated with activation of myeloid cells such as microglia and macrophages. The chemokine fractalkine (FKN or Cx3cl1) mediates communication from neurons to myeloid cells. Signaling through its receptor Cx3cr1 has been implicated in multiple neurodegenerative diseases, but the effects on neuronal pathology are variable. Since it is unknown how FKN-mediated crosstalk influences RGC degeneration in glaucoma, we assessed this in a chronic mouse model, DBA/2J. We analyzed a DBA/2J substrain deficient in Cx3cr1, and compared compartmentalized RGC degeneration and myeloid cell responses to those in standard DBA/2J mice. We found that loss of FKN signaling exacerbates axon transport dysfunction, an early event in neurodegeneration, with a significant increase in RGCs with somal accumulation of the axonal protein phosphorylated neurofilament, and reduced retinal expression of genes involved in axon transport, Kif1b, and Atp8a2. There was no change in the loss of Brn3-positive RGCs, and no difference in the extent of damage to the proximal optic nerve, suggesting that the loss of fractalkine signaling primarily affects axon transport. Since Cx3cr1 is specifically expressed in myeloid cells, we assessed changes in retinal microglial number and activation, changes in gene expression, and the extent of macrophage infiltration. We found that loss of fractalkine signaling led to innate immune changes within the retina, including increased infiltration of peripheral macrophages and upregulated nitric oxide synthase-2 (Nos-2) expression in myeloid cells, which contributes to the production of NO and can promote axon transport deficits. In contrast, resident retinal microglia appeared unchanged either in number, morphology, or expression of the myeloid activation marker ionized calcium binding adaptor molecule 1 (Iba1). There was also no significant increase in the proinflammatory gene interleukin 1 beta (Il1β). We conclude that loss of fractalkine signaling causes a selective worsening of axon transport dysfunction in RGCs, which is linked to enhanced Nos-2 expression in myeloid cells. Our findings suggest that distinct mechanisms may contribute to different aspects of RGC decline in glaucoma, with axonal transport selectively altered after loss of Cx3cr1 in microglia and/or macrophages.
Collapse
Affiliation(s)
- Kevin T Breen
- Departments of Neurobiology and Anatomy, University of Utah Salt Lake City, UT, USA
| | - Sarah R Anderson
- Departments of Neurobiology and Anatomy, University of Utah Salt Lake City, UT, USA
| | - Michael R Steele
- Departments of Neurobiology and Anatomy, University of Utah Salt Lake City, UT, USA
| | - David J Calkins
- Department of Ophthalmology and Visual Sciences, Vanderbilt University Nashville, TN, USA
| | - Alejandra Bosco
- Departments of Neurobiology and Anatomy, University of Utah Salt Lake City, UT, USA
| | - Monica L Vetter
- Departments of Neurobiology and Anatomy, University of Utah Salt Lake City, UT, USA
| |
Collapse
|
22
|
Progesterone Attenuates Microglial-Driven Retinal Degeneration and Stimulates Protective Fractalkine-CX3CR1 Signaling. PLoS One 2016; 11:e0165197. [PMID: 27814376 PMCID: PMC5096718 DOI: 10.1371/journal.pone.0165197] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 10/08/2016] [Indexed: 01/29/2023] Open
Abstract
Retinitis pigmentosa (RP) is a degenerative disease leading to photoreceptor cell loss. Mouse models of RP, such as the rd10 mouse (B6.CXBl-Pde6brd10/J), have enhanced our understanding of the disease, allowing for development of potential therapeutics. In 2011, our group first demonstrated that the synthetic progesterone analogue ‘Norgestrel’ is neuroprotective in two mouse models of retinal degeneration, including the rd10 mouse. We have since elucidated several mechanisms by which Norgestrel protects stressed photoreceptors, such as upregulating growth factors. This study consequently aimed to further characterize Norgestrel’s neuroprotective effects. Specifically, we sought to investigate the role that microglia might play; for microglial-derived inflammation has been shown to potentiate neurodegeneration. Dams of post-natal day (P) 10 rd10 pups were given a Norgestrel-supplemented diet (80mg/kg). Upon weaning, pups remained on Norgestrel. Tissue was harvested from P15-P50 rd10 mice on control or Norgestrel-supplemented diet. Norgestrel-diet administration provided significant retinal protection out to P40 in rd10 mice. Alterations in microglial activity coincided with significant protection, implicating microglial changes in Norgestrel-induced neuroprotection. Utilizing primary cultures of retinal microglia and 661W photoreceptor-like cells, we show that rd10 microglia drive neuronal cell death. We reveal a novel role of Norgestrel, acting directly on microglia to reduce pro-inflammatory activation and prevent neuronal cell death. Norgestrel effectively suppresses cytokine, chemokine and danger-associated molecular pattern molecule (DAMP) expression in the rd10 retina. Remarkably, Norgestrel upregulates fractalkine-CX3CR1 signaling 1 000-fold at the RNA level, in the rd10 mouse. Fractalkine-CX3CR1 signaling has been shown to protect neurons by regulating retinal microglial activation and migration. Ultimately, these results present Norgestrel as a promising treatment for RP, with dual actions as a neuroprotective and anti-inflammatory agent in the retina.
Collapse
|
23
|
Beli E, Dominguez JM, Hu P, Thinschmidt JS, Caballero S, Li Calzi S, Luo D, Shanmugam S, Salazar TE, Duan Y, Boulton ME, Mohr S, Abcouwer SF, Saban DR, Harrison JK, Grant MB. CX3CR1 deficiency accelerates the development of retinopathy in a rodent model of type 1 diabetes. J Mol Med (Berl) 2016; 94:1255-1265. [PMID: 27344677 PMCID: PMC5071129 DOI: 10.1007/s00109-016-1433-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2015] [Revised: 05/17/2016] [Accepted: 05/25/2016] [Indexed: 12/31/2022]
Abstract
In this study, the role of CX3CR1 in the progression of diabetic retinopathy (DR) was investigated. The retinas of wild-type (WT), CX3CR1 null (CX3CR1gfp/gfp, KO), and heterozygous (CX3CR1+/gfp, Het) mice were compared in the presence and absence of streptozotocin (STZ)-induced diabetes. CX3CR1 deficiency in STZ-KO increased vascular pathology at 4 months of diabetes, as a significant increase in acellular capillaries was observed only in the STZ-KO group. CX3CR1 deficiency and diabetes had similar effects on retinal neurodegeneration measured by an increase in DNA fragmentation. Retinal vascular pathology in STZ-KO mice was associated with increased numbers of monocyte-derived macrophages in the retina. Furthermore, compared to STZ-WT, STZ-KO mice exhibited increased numbers of inflammatory monocytes in the bone marrow and impaired homing of monocytes to the spleen. The induction of retinal IL-10 expression by diabetes was significantly less in KO mice, and when bone marrow-derived macrophages from KO mice were maintained in high glucose, they expressed significantly less IL-10 and more TNF-α in response to LPS stimulation. These findings support that CX3CR1 deficiency accelerates the development of vascular pathology in DR through increased recruitment of proinflammatory myeloid cells that demonstrate reduced expression of anti-inflammatory IL-10. KEY MESSAGES • CX3CR1 deletion in STZ-diabetic mice accelerated the onset of diabetic retinopathy (DR). • The early onset of DR was associated with increased retinal cell apoptosis. • The early onset of DR was associated with increased recruitment of bone marrow-derived macrophages to the retina. • Bone marrow-derived macrophages from CX3CR1 KO diabetic mice expressed more TNF-α and less IL-10. • The role of IL-10 in protection from progression of DR is highlighted.
Collapse
Affiliation(s)
- Eleni Beli
- Eugene and Marilyn Glick Eye Institute, Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - James M Dominguez
- Eugene and Marilyn Glick Eye Institute, Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, USA
| | - Ping Hu
- Eugene and Marilyn Glick Eye Institute, Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jeffrey S Thinschmidt
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, USA
| | - Sergio Caballero
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, USA
| | - Sergio Li Calzi
- Eugene and Marilyn Glick Eye Institute, Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Defang Luo
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, USA
| | - Sumathi Shanmugam
- Department of Ophthalmology and Visual Sciences, University of Michigan, Kellogg Eye Center, Ann Arbor, MI, USA
| | - Tatiana E Salazar
- Eugene and Marilyn Glick Eye Institute, Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, USA
| | - Yaqian Duan
- Eugene and Marilyn Glick Eye Institute, Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Michael E Boulton
- Eugene and Marilyn Glick Eye Institute, Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Susanna Mohr
- Department of Physiology, Michigan State University, East Lancing, MI, USA
| | - Steven F Abcouwer
- Department of Ophthalmology and Visual Sciences, University of Michigan, Kellogg Eye Center, Ann Arbor, MI, USA
| | - Daniel R Saban
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, USA
| | - Jeffrey K Harrison
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, USA
| | - Maria B Grant
- Eugene and Marilyn Glick Eye Institute, Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN, USA.
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
24
|
Eandi CM, Charles Messance H, Augustin S, Dominguez E, Lavalette S, Forster V, Hu SJ, Siquieros L, Craft CM, Sahel JA, Tadayoni R, Paques M, Guillonneau X, Sennlaub F. Subretinal mononuclear phagocytes induce cone segment loss via IL-1β. eLife 2016; 5. [PMID: 27438413 PMCID: PMC4969036 DOI: 10.7554/elife.16490] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Accepted: 07/19/2016] [Indexed: 12/19/2022] Open
Abstract
Photo-transduction in cone segments (CS) is crucial for high acuity daytime vision. For ill-defined reasons, CS degenerate in retinitis pigmentosa (RP) and in the transitional zone (TZ) of atrophic zones (AZ), which characterize geographic atrophy (GA). Our experiments confirm the loss of cone segments (CS) in the TZ of patients with GA and show their association with subretinal CD14+mononuclear phagocyte (MP) infiltration that is also reported in RP. Using human and mouse MPs in vitro and inflammation-prone Cx3cr1GFP/GFPmice in vivo, we demonstrate that MP-derived IL-1β leads to severe CS degeneration. Our results strongly suggest that subretinal MP accumulation participates in the observed pathological photoreceptor changes in these diseases. Inhibiting subretinal MP accumulation or Il-1β might protect the CS and help preserve high acuity daytime vision in conditions characterized by subretinal inflammation, such as AMD and RP. DOI:http://dx.doi.org/10.7554/eLife.16490.001
Collapse
Affiliation(s)
- Chiara M Eandi
- Sorbonne Universités, UPMC University Paris 06, INSERM, CNRS, Paris, France.,Institut de la Vision, Paris, France.,Department of Clinical Science, University of Torino, Torino, Italy.,Eye Clinic, University of Torino, Torino, Italy
| | - Hugo Charles Messance
- Sorbonne Universités, UPMC University Paris 06, INSERM, CNRS, Paris, France.,Institut de la Vision, Paris, France
| | - Sébastien Augustin
- Sorbonne Universités, UPMC University Paris 06, INSERM, CNRS, Paris, France.,Institut de la Vision, Paris, France
| | - Elisa Dominguez
- Sorbonne Universités, UPMC University Paris 06, INSERM, CNRS, Paris, France.,Institut de la Vision, Paris, France
| | - Sophie Lavalette
- Sorbonne Universités, UPMC University Paris 06, INSERM, CNRS, Paris, France.,Institut de la Vision, Paris, France
| | - Valérie Forster
- Sorbonne Universités, UPMC University Paris 06, INSERM, CNRS, Paris, France.,Institut de la Vision, Paris, France
| | - Shulong Justin Hu
- Sorbonne Universités, UPMC University Paris 06, INSERM, CNRS, Paris, France.,Institut de la Vision, Paris, France
| | - Lourdes Siquieros
- Sorbonne Universités, UPMC University Paris 06, INSERM, CNRS, Paris, France.,Institut de la Vision, Paris, France
| | - Cheryl Mae Craft
- Mary D. Allen Laboratory for Vision Research, Keck School of Medicine of the University of Southern California, Los Angeles, United States.,University of Southern California Eye Institute, Keck School of Medicine of the University of Southern California, Los Angeles, United States.,Department of Ophthalmology and Cell, Keck School of Medicine of the University of Southern California, Los Angeles, United States.,Department of Neurobiology, Keck School of Medicine of the University of Southern California, Los Angeles, United States
| | - José-Alain Sahel
- Sorbonne Universités, UPMC University Paris 06, INSERM, CNRS, Paris, France.,Institut de la Vision, Paris, France.,Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, INSERM-DHOS CIC 503, Paris, France
| | - Ramin Tadayoni
- Sorbonne Universités, UPMC University Paris 06, INSERM, CNRS, Paris, France.,Institut de la Vision, Paris, France.,Department of Ophthalmology, Hôpital Lariboisièr, Paris, France
| | - Michel Paques
- Sorbonne Universités, UPMC University Paris 06, INSERM, CNRS, Paris, France.,Institut de la Vision, Paris, France.,Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, INSERM-DHOS CIC 503, Paris, France
| | - Xavier Guillonneau
- Sorbonne Universités, UPMC University Paris 06, INSERM, CNRS, Paris, France.,Institut de la Vision, Paris, France
| | - Florian Sennlaub
- Sorbonne Universités, UPMC University Paris 06, INSERM, CNRS, Paris, France.,Institut de la Vision, Paris, France
| |
Collapse
|
25
|
Zabel MK, Zhao L, Zhang Y, Gonzalez SR, Ma W, Wang X, Fariss RN, Wong WT. Microglial phagocytosis and activation underlying photoreceptor degeneration is regulated by CX3CL1-CX3CR1 signaling in a mouse model of retinitis pigmentosa. Glia 2016; 64:1479-91. [PMID: 27314452 PMCID: PMC4958518 DOI: 10.1002/glia.23016] [Citation(s) in RCA: 130] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Accepted: 05/20/2016] [Indexed: 12/20/2022]
Abstract
Retinitis pigmentosa (RP), a disease characterized by the progressive degeneration of mutation‐bearing photoreceptors, is a significant cause of incurable blindness in the young worldwide. Recent studies have found that activated retinal microglia contribute to photoreceptor demise via phagocytosis and proinflammatory factor production, however mechanisms regulating these contributions are not well‐defined. In this study, we investigate the role of CX3CR1, a microglia‐specific receptor, in regulating microglia‐mediated degeneration using the well‐established rd10 mouse model of RP. We found that in CX3CR1‐deficient (CX3CR1GFP/GFP) rd10 mice microglial infiltration into the photoreceptor layer was significantly augmented and associated with accelerated photoreceptor apoptosis and atrophy compared with CX3CR1‐sufficient (CX3CR1GFP/+) rd10 littermates. CX3CR1‐deficient microglia demonstrated increased phagocytosis as evidenced by (1) having increased numbers of phagosomes in vivo, (2) an increased rate of phagocytosis of fluorescent beads and photoreceptor cellular debris in vitro, and (3) increased photoreceptor phagocytosis dynamics on live cell imaging in retinal explants, indicating that CX3CR1 signaling in microglia regulates the phagocytic clearance of at‐risk photoreceptors. We also found that CX3CR1 deficiency in retinal microglia was associated with increased expression of inflammatory cytokines and microglial activation markers. Significantly, increasing CX3CL1‐CX3CR1 signaling in the rd10 retina via exogenous intravitreal delivery of recombinant CX3CL1 was effective in (1) decreasing microglial infiltration, phagocytosis and activation, and (2) improving structural and functional features of photoreceptor degeneration. These results indicate that CX3CL1‐CX3CR1 signaling is a molecular mechanism capable of modulating microglial‐mediated degeneration and represents a potential molecular target in therapeutic approaches to RP. GLIA 2016;64:1479–1491
Collapse
Affiliation(s)
- Matthew K Zabel
- Unit on Neuron-Glia Interactions in Retinal Disease, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Lian Zhao
- Unit on Neuron-Glia Interactions in Retinal Disease, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Yikui Zhang
- Unit on Neuron-Glia Interactions in Retinal Disease, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Shaimar R Gonzalez
- Unit on Neuron-Glia Interactions in Retinal Disease, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Wenxin Ma
- Unit on Neuron-Glia Interactions in Retinal Disease, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Xu Wang
- Unit on Neuron-Glia Interactions in Retinal Disease, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Robert N Fariss
- Biological Imaging Core, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Wai T Wong
- Unit on Neuron-Glia Interactions in Retinal Disease, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
26
|
Greene JA, Portillo JAC, Lopez Corcino Y, Subauste CS. CD40-TRAF Signaling Upregulates CX3CL1 and TNF-α in Human Aortic Endothelial Cells but Not in Retinal Endothelial Cells. PLoS One 2015; 10:e0144133. [PMID: 26710229 PMCID: PMC4692437 DOI: 10.1371/journal.pone.0144133] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2015] [Accepted: 11/14/2015] [Indexed: 11/23/2022] Open
Abstract
CD40, CX3CL1 and TNF-α promote atheroma and neointima formation. CD40 and TNF-α are also central to the development of diabetic retinopathy while CX3CL1 may play a role in the pathogenesis of this retinopathy. The purpose of this study was to examine whether CD40 ligation increases CX3CL1 and TNF-α protein expression in human endothelial cells from the aorta and retina. CD154 (CD40 ligand) upregulated membrane-bound and soluble CX3CL1 in human aortic endothelial cells. CD154 triggered TNF-α production by human aortic endothelial cells. TNF Receptor Associated Factors (TRAF) are key mediators of CD40 signaling. Compared to human aortic endothelial cells that express wt CD40, cells that express CD40 with a mutation that prevents TRAF2,3 recruitment, or CD40 with a mutation that prevents TRAF6 recruitment exhibited a profound inhibition of CD154-driven upregulation of membrane bound and soluble CX3CL1 as well as of TNF-α secretion. While both CD154 and TNF-α upregulated CX3CL1 in human aortic endothelial cells, these stimuli could act independently of each other. In contrast to human aortic endothelial cells, human retinal endothelial cells did not increase membrane bound or soluble CX3CL1 expression or secrete TNF-α in response to CD154 even though CD40 ligation upregulated ICAM-1 and CCL2 in these cells. Moreover, TNF-α did not upregulate CX3CL1 in retinal endothelial cells. In conclusion, CD40 ligation increases CX3CL1 protein levels and induces TNF-α production in endothelial cells. However, endothelial cells are heterogeneous in regards to these responses. Human aortic but not retinal endothelial cells upregulated CX3CL1 and TNF-α in response to CD40 ligation, as well as upregulated CX3CL1 in response to TNF-α. These dissimilarities may contribute to differences in regulation of inflammation in large vessels versus the retina.
Collapse
Affiliation(s)
- Jennifer A. Greene
- Division of Infectious Diseases and HIV Medicine, Dept. of Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Jose-Andres C. Portillo
- Division of Infectious Diseases and HIV Medicine, Dept. of Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Yalitza Lopez Corcino
- Division of Infectious Diseases and HIV Medicine, Dept. of Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Carlos S. Subauste
- Division of Infectious Diseases and HIV Medicine, Dept. of Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, Ohio, United States of America
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, United States of America
- * E-mail:
| |
Collapse
|
27
|
Levy O, Calippe B, Lavalette S, Hu SJ, Raoul W, Dominguez E, Housset M, Paques M, Sahel JA, Bemelmans AP, Combadiere C, Guillonneau X, Sennlaub F. Apolipoprotein E promotes subretinal mononuclear phagocyte survival and chronic inflammation in age-related macular degeneration. EMBO Mol Med 2015; 7:211-26. [PMID: 25604058 PMCID: PMC4328649 DOI: 10.15252/emmm.201404524] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Physiologically, the retinal pigment epithelium (RPE) expresses immunosuppressive signals such as FAS ligand (FASL), which prevents the accumulation of leukocytes in the subretinal space. Age-related macular degeneration (AMD) is associated with a breakdown of the subretinal immunosuppressive environment and chronic accumulation of mononuclear phagocytes (MPs). We show that subretinal MPs in AMD patients accumulate on the RPE and express high levels of APOE. MPs of Cx3cr1(-/-) mice that develop MP accumulation on the RPE, photoreceptor degeneration, and increased choroidal neovascularization similarly express high levels of APOE. ApoE deletion in Cx3cr1(-/-) mice prevents pathogenic age- and stress-induced subretinal MP accumulation. We demonstrate that increased APOE levels induce IL-6 in MPs via the activation of the TLR2-CD14-dependent innate immunity receptor cluster. IL-6 in turn represses RPE FasL expression and prolongs subretinal MP survival. This mechanism may account, in part, for the MP accumulation observed in Cx3cr1(-/-) mice. Our results underline the inflammatory role of APOE in sterile inflammation in the immunosuppressive subretinal space. They provide rationale for the implication of IL-6 in AMD and open avenues toward therapies inhibiting pathogenic chronic inflammation in late AMD.
Collapse
Affiliation(s)
- Olivier Levy
- INSERM, Paris, France UPMC Univ Paris 06 UMR_S 968 Institut de la Vision, Paris, France Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts INSERM-DHOS CIC 503, Paris, France
| | - Bertrand Calippe
- INSERM, Paris, France UPMC Univ Paris 06 UMR_S 968 Institut de la Vision, Paris, France Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts INSERM-DHOS CIC 503, Paris, France
| | - Sophie Lavalette
- INSERM, Paris, France UPMC Univ Paris 06 UMR_S 968 Institut de la Vision, Paris, France Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts INSERM-DHOS CIC 503, Paris, France
| | - Shulong J Hu
- INSERM, Paris, France UPMC Univ Paris 06 UMR_S 968 Institut de la Vision, Paris, France Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts INSERM-DHOS CIC 503, Paris, France
| | - William Raoul
- INSERM, Paris, France UPMC Univ Paris 06 UMR_S 968 Institut de la Vision, Paris, France Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts INSERM-DHOS CIC 503, Paris, France
| | - Elisa Dominguez
- INSERM, Paris, France UPMC Univ Paris 06 UMR_S 968 Institut de la Vision, Paris, France Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts INSERM-DHOS CIC 503, Paris, France
| | - Michael Housset
- INSERM, Paris, France UPMC Univ Paris 06 UMR_S 968 Institut de la Vision, Paris, France Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts INSERM-DHOS CIC 503, Paris, France
| | - Michel Paques
- INSERM, Paris, France UPMC Univ Paris 06 UMR_S 968 Institut de la Vision, Paris, France Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts INSERM-DHOS CIC 503, Paris, France
| | - José-Alain Sahel
- INSERM, Paris, France UPMC Univ Paris 06 UMR_S 968 Institut de la Vision, Paris, France Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts INSERM-DHOS CIC 503, Paris, France
| | - Alexis-Pierre Bemelmans
- INSERM, Paris, France UPMC Univ Paris 06 UMR_S 968 Institut de la Vision, Paris, France Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts INSERM-DHOS CIC 503, Paris, France CEA DSV I²BM Molecular Imaging Research Center (MIRCen), Fontenay-aux-Roses, France CNRS CEA URA 2210, Fontenay-aux-Roses, France
| | - Christophe Combadiere
- Sorbonne Universités, UPMC Univ Paris 06 CR7 Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France INSERM U1135 CIMI-Paris, Paris, France CNRS ERL 8255 CIMI-Paris, Paris, France
| | - Xavier Guillonneau
- INSERM, Paris, France UPMC Univ Paris 06 UMR_S 968 Institut de la Vision, Paris, France Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts INSERM-DHOS CIC 503, Paris, France
| | - Florian Sennlaub
- INSERM, Paris, France UPMC Univ Paris 06 UMR_S 968 Institut de la Vision, Paris, France Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts INSERM-DHOS CIC 503, Paris, France
| |
Collapse
|
28
|
Upregulation of P2RX7 in Cx3cr1-Deficient Mononuclear Phagocytes Leads to Increased Interleukin-1β Secretion and Photoreceptor Neurodegeneration. J Neurosci 2015; 35:6987-96. [PMID: 25948251 DOI: 10.1523/jneurosci.3955-14.2015] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Photoreceptor degeneration in age-related macular degeneration (AMD) is associated with an infiltration and chronic accumulation of mononuclear phagocytes (MPs). We have previously shown that Cx3cr1-deficient mice develop age- and stress- related subretinal accumulation of MPs, which is associated with photoreceptor degeneration. Cx3cr1-deficient MPs have been shown to increase neuronal apoptosis through IL-1β in neuroinflammation of the brain. The reason for increased IL-1β secretion from Cx3cr1-deficient MPs, and whether IL-1β is responsible for increased photoreceptor apoptosis in Cx3cr1-deficient mice, has not been elucidated. Here we show that Cx3cr1-deficient MPs express increased surface P2X7 receptor (P2RX7), which stimulates IL-1β maturation and secretion. P2RX7 and IL-1β inhibition efficiently blunted Cx3cr1-MP-dependent photoreceptor apoptosis in a monocyte/retina coculture system and in light-induced subretinal inflammation of Cx3cr1-deficient mice in vivo. Our results provide an explanation for increased CX3CR1-dependent IL-1β secretion and suggest that IL-1β or P2RX7 inhibition can help inhibit the inflammation-associated photoreceptor cell loss in late AMD, including geographic atrophy, for which no efficient treatment currently exists.
Collapse
|
29
|
Vecino E, Rodriguez FD, Ruzafa N, Pereiro X, Sharma SC. Glia-neuron interactions in the mammalian retina. Prog Retin Eye Res 2015; 51:1-40. [PMID: 26113209 DOI: 10.1016/j.preteyeres.2015.06.003] [Citation(s) in RCA: 553] [Impact Index Per Article: 55.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 05/18/2015] [Accepted: 06/02/2015] [Indexed: 02/07/2023]
Abstract
The mammalian retina provides an excellent opportunity to study glia-neuron interactions and the interactions of glia with blood vessels. Three main types of glial cells are found in the mammalian retina that serve to maintain retinal homeostasis: astrocytes, Müller cells and resident microglia. Müller cells, astrocytes and microglia not only provide structural support but they are also involved in metabolism, the phagocytosis of neuronal debris, the release of certain transmitters and trophic factors and K(+) uptake. Astrocytes are mostly located in the nerve fibre layer and they accompany the blood vessels in the inner nuclear layer. Indeed, like Müller cells, astrocytic processes cover the blood vessels forming the retinal blood barrier and they fulfil a significant role in ion homeostasis. Among other activities, microglia can be stimulated to fulfil a macrophage function, as well as to interact with other glial cells and neurons by secreting growth factors. This review summarizes the main functional relationships between retinal glial cells and neurons, presenting a general picture of the retina recently modified based on experimental observations. The preferential involvement of the distinct glia cells in terms of the activity in the retina is discussed, for example, while Müller cells may serve as progenitors of retinal neurons, astrocytes and microglia are responsible for synaptic pruning. Since different types of glia participate together in certain activities in the retina, it is imperative to explore the order of redundancy and to explore the heterogeneity among these cells. Recent studies revealed the association of glia cell heterogeneity with specific functions. Finally, the neuroprotective effects of glia on photoreceptors and ganglion cells under normal and adverse conditions will also be explored.
Collapse
Affiliation(s)
- Elena Vecino
- Department of Cell Biology and Histology, University of the Basque Country UPV/EHU, Leioa 48940, Vizcaya, Spain
| | - F David Rodriguez
- Department of Biochemistry and Molecular Biology, E-37007, University of Salamanca, Salamanca, Spain
| | - Noelia Ruzafa
- Department of Cell Biology and Histology, University of the Basque Country UPV/EHU, Leioa 48940, Vizcaya, Spain
| | - Xandra Pereiro
- Department of Cell Biology and Histology, University of the Basque Country UPV/EHU, Leioa 48940, Vizcaya, Spain
| | - Sansar C Sharma
- Department of Ophthalmology, Cell Biology and Anatomy, New York Medical College, Valhalla, NY 10595, USA; IKERBASQUE, Basque Foundation for Science at Dept. Cell Biology and Histology, UPV/EHU, Spain
| |
Collapse
|