1
|
Biber J, Jabri Y, Glänzer S, Dort A, Hoffelner P, Schmidt CQ, Bludau O, Pauly D, Grosche A. Gliosis-dependent expression of complement factor H truncated variants attenuates retinal neurodegeneration following ischemic injury. J Neuroinflammation 2024; 21:56. [PMID: 38388518 PMCID: PMC10885619 DOI: 10.1186/s12974-024-03045-3] [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: 10/11/2023] [Accepted: 02/14/2024] [Indexed: 02/24/2024] Open
Abstract
Inherited, age-related, and acute retinal diseases are often exacerbated by an aberrant or excessive activity of the complement system. Consequently, cells not directly affected by an acute event or genetic variants may degenerate, resulting in enhanced visual impairment. The therapeutic potential of supplementation of complement factor H (FH), a key regulator of the complement cascade, is therefore particularly promising in the context of retinal diseases caused by complement activation. In this study, we engineered adeno-associated viruses (AAVs) containing sequences of two truncated human FH variants. The expression of these variants was regulated by the glial fibrillary acidic protein (GFAP) promoter, which is selectively active in gliotic Müller cells. Both FH variants consisted of FH domains 19-20, which were connected to domains 1-4 and 1-7, respectively, by a polyglycine linker. These AAVs were intravitreally injected following ischemic injury of C57BL/6J mouse retinas. We observed transgene expression in gliotic Müller cells and to some extent in astrocytes. The expression correlated directly with damage severity. Interventions resulted in decreased complement activation, accelerated normalization of microglia activity and morphological improvements. Reduced levels of C3 transcripts and C3d protein in conjunction with higher transcript levels of inhibitory regulators like Cfi and Cfh, hinted at attenuated complement activity. This study demonstrates the great potential of complement regulatory gene addition therapy. With further in vivo testing it could be applied to treat a wide range of retinal diseases where no causative therapies are available.
Collapse
Affiliation(s)
- Josef Biber
- Department of Physiological Genomics, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany
| | - Yassin Jabri
- Department of Ophthalmology, University Hospital Regensburg, Regensburg, Germany
| | - Sarah Glänzer
- Department of Physiological Genomics, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany
| | - Aaron Dort
- Experimental Ophthalmology, University of Marburg, Marburg, Germany
| | - Patricia Hoffelner
- Department of Physiological Genomics, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany
| | - Christoph Q Schmidt
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, University of Ulm Medical Center, Ulm, Germany
- Institute of Pharmacy, Biochemical Pharmacy Group, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Oliver Bludau
- Department of Physiological Genomics, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany
| | - Diana Pauly
- Experimental Ophthalmology, University of Marburg, Marburg, Germany.
| | - Antje Grosche
- Department of Physiological Genomics, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany
| |
Collapse
|
2
|
Pfaller AM, Kaplan L, Carido M, Grassmann F, Díaz-Lezama N, Ghaseminejad F, Wunderlich KA, Glänzer S, Bludau O, Pannicke T, Weber BHF, Koch SF, Bonev B, Hauck SM, Grosche A. The glucocorticoid receptor as a master regulator of the Müller cell response to diabetic conditions in mice. J Neuroinflammation 2024; 21:33. [PMID: 38273366 PMCID: PMC10809506 DOI: 10.1186/s12974-024-03021-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 01/11/2024] [Indexed: 01/27/2024] Open
Abstract
Diabetic retinopathy (DR) is considered a primarily microvascular complication of diabetes. Müller glia cells are at the centre of the retinal neurovascular unit and play a critical role in DR. We therefore investigated Müller cell-specific signalling pathways that are altered in DR to identify novel targets for gene therapy. Using a multi-omics approach on purified Müller cells from diabetic db/db mice, we found the mRNA and protein expression of the glucocorticoid receptor (GR) to be significantly decreased, while its target gene cluster was down-regulated. Further, oPOSSUM TF analysis and ATAC- sequencing identified the GR as a master regulator of Müller cell response to diabetic conditions. Cortisol not only increased GR phosphorylation. It also induced changes in the expression of known GR target genes in retinal explants. Finally, retinal functionality was improved by AAV-mediated overexpression of GR in Müller cells. Our study demonstrates an important role of the glial GR in DR and implies that therapeutic approaches targeting this signalling pathway should be aimed at increasing GR expression rather than the addition of more ligand.
Collapse
Affiliation(s)
- Anna M Pfaller
- Department of Physiological Genomics, Biomedical Center-BMC, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany
| | - Lew Kaplan
- Department of Physiological Genomics, Biomedical Center-BMC, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany
| | - Madalena Carido
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Felix Grassmann
- Institute of Clinical Human Genetics, University Hospital Regensburg, Regensburg, Germany
- Institute for Clinical Research and Systems Medicine, Health and Medical University, Potsdam, Germany
| | - Nundehui Díaz-Lezama
- Department of Physiological Genomics, Biomedical Center-BMC, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Farhad Ghaseminejad
- Department of Physiological Genomics, Biomedical Center-BMC, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany
| | - Kirsten A Wunderlich
- Department of Physiological Genomics, Biomedical Center-BMC, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany
- Institute for Molecular Medicine, Health and Medical University, Potsdam, Germany
| | - Sarah Glänzer
- Department of Physiological Genomics, Biomedical Center-BMC, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany
| | - Oliver Bludau
- Department of Physiological Genomics, Biomedical Center-BMC, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany
| | - Thomas Pannicke
- Paul Flechsig Institute for Brain Research, University of Leipzig, Leipzig, Germany
| | - Bernhard H F Weber
- Institute of Clinical Human Genetics, University Hospital Regensburg, Regensburg, Germany
- Institute of Human Genetics, University Regensburg, Regensburg, Germany
| | - Susanne F Koch
- Department of Physiological Genomics, Biomedical Center-BMC, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Boyan Bonev
- Department of Physiological Genomics, Biomedical Center-BMC, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Stefanie M Hauck
- Metabolomics and Proteomics Core, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Antje Grosche
- Department of Physiological Genomics, Biomedical Center-BMC, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany.
| |
Collapse
|
3
|
Daruich A, Robert MP, Zola M, Matet A, Bremond-Gignac D. Retinal stroke: research models, targets and experimental drugs. Expert Opin Investig Drugs 2023; 32:755-760. [PMID: 37651742 DOI: 10.1080/13543784.2023.2254688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 08/02/2023] [Accepted: 08/30/2023] [Indexed: 09/02/2023]
Abstract
INTRODUCTION Retinal artery occlusion (RAO), often caused by a microembolus and resulting in inner retinal ischemia, could be considered as the retinal analog to cerebral stroke. Although several therapeutic targets have been suggested in animal models of retinal ischemia and several potential treatments have been evaluated on small series of patients, central retinal artery occlusion (CRAO) is still rarely treatable in clinical practice. AREAS COVERED Here, we review several animal models of RAO, including increased intraocular pressure, laser, vasoconstriction, embolization and clamp. We also review the pathogenic mechanisms that contribute to cell death cascades during ischemia, and the therapeutic strategies targeting these events. These strategies aim to restore blood flow by fibrinolysis, increase the oxygen or glucose supply, decrease the energy demands, restrict ionic leak fluxes or reduce the detrimental effects of glutamate, calcium and free radicals. The current literature suggests that tPA treatment could be effective for CRAO. EXPERT OPINION Eye care professionals must make a rapid and accurate diagnosis and immediately refer patients with acute retinal stroke to specialized centers. CRAO management should also be facilitated by developing local networks to encourage collaboration among ophthalmologists, retina specialists and stroke neurologists.
Collapse
Affiliation(s)
- Alejandra Daruich
- Ophthalmology Department, Necker-Enfants Malades University Hospital, AP-HP, Paris Cité University, Paris, France
- INSERM, UMRS1138, Team 17, From physiopathology of ocular diseases to clinical development, Sorbonne Paris Cité University, Paris, France
| | - Matthieu P Robert
- Ophthalmology Department, Necker-Enfants Malades University Hospital, AP-HP, Paris Cité University, Paris, France
- Borelli Centre, UMR 9010, CNRS-SSA-ENS Paris Saclay-Paris Cité University, Paris, France
| | - Marta Zola
- INSERM, UMRS1138, Team 17, From physiopathology of ocular diseases to clinical development, Sorbonne Paris Cité University, Paris, France
| | - Alexandre Matet
- Ophthalmology Department, Institut Curie, Paris Cité University, Paris, France
| | - Dominique Bremond-Gignac
- Ophthalmology Department, Necker-Enfants Malades University Hospital, AP-HP, Paris Cité University, Paris, France
- INSERM, UMRS1138, Team 17, From physiopathology of ocular diseases to clinical development, Sorbonne Paris Cité University, Paris, France
| |
Collapse
|
4
|
Miao Y, Zhao GL, Cheng S, Wang Z, Yang XL. Activation of retinal glial cells contributes to the degeneration of ganglion cells in experimental glaucoma. Prog Retin Eye Res 2023; 93:101169. [PMID: 36736070 DOI: 10.1016/j.preteyeres.2023.101169] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 01/12/2023] [Accepted: 01/24/2023] [Indexed: 02/04/2023]
Abstract
Elevation of intraocular pressure (IOP) is a major risk factor for neurodegeneration in glaucoma. Glial cells, which play an important role in normal functioning of retinal neurons, are well involved into retinal ganglion cell (RGC) degeneration in experimental glaucoma animal models generated by elevated IOP. In response to elevated IOP, mGluR I is first activated and Kir4.1 channels are subsequently inhibited, which leads to the activation of Müller cells. Müller cell activation is followed by a complex process, including proliferation, release of inflammatory and growth factors (gliosis). Gliosis is further regulated by several factors. Activated Müller cells contribute to RGC degeneration through generating glutamate receptor-mediated excitotoxicity, releasing cytotoxic factors and inducing microglia activation. Elevated IOP activates microglia, and following morphological and functional changes, these cells, as resident immune cells in the retina, show adaptive immune responses, including an enhanced release of pro-inflammatory factors (tumor neurosis factor-α, interleukins, etc.). These ATP and Toll-like receptor-mediated responses are further regulated by heat shock proteins, CD200R, chemokine receptors, and metabotropic purinergic receptors, may aggravate RGC loss. In the optic nerve head, astrogliosis is initiated and regulated by a complex reaction process, including purines, transmitters, chemokines, growth factors and cytokines, which contributes to RGC axon injury through releasing pro-inflammatory factors and changing extracellular matrix in glaucoma. The effects of activated glial cells on RGCs are further modified by the interplay among different types of glial cells. This review is concluded by presenting an in-depth discussion of possible research directions in this field in the future.
Collapse
Affiliation(s)
- Yanying Miao
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Guo-Li Zhao
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Shuo Cheng
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Zhongfeng Wang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China.
| | - Xiong-Li Yang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
5
|
Shinozaki Y, Saito K, Kashiwagi K, Koizumi S. Ocular P2 receptors and glaucoma. Neuropharmacology 2023; 222:109302. [PMID: 36341810 DOI: 10.1016/j.neuropharm.2022.109302] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 10/08/2022] [Accepted: 10/21/2022] [Indexed: 11/06/2022]
Abstract
Adenosine triphosphate (ATP), an energy source currency in cells, is released or leaked to the extracellular space under both physiological and pathological conditions. Extracellular ATP functions as an intercellular signaling molecule through activation of purinergic P2 receptors. Ocular tissue and cells release ATP in response to physiological stimuli such as intraocular pressure (IOP), and P2 receptor activation regulates IOP elevation or reduction. Dysregulated purinergic signaling may cause abnormally elevated IOP, which is one of the major risk factors for glaucoma. Glaucoma, a leading cause of blindness worldwide, is characterized by progressive degeneration of optic nerves and retinal ganglion cells (RGCs), which are essential retinal neurons that transduce visual information to the brain. An elevation in IOP may stress RGCs and increase the risk for glaucoma pathogenesis. In the aqueous humor of human patients with glaucoma, the ATP level is significantly elevated. Such excess amount of ATP may directly cause RGC death via a specific subtype of P2 receptors. Dysregulated purinergic signaling may also trigger inflammation, oxidative stress, and excitotoxicity via activating non-neuronal cell types such as glial cells. In this review, we discussed the physiological roles of extracellular nucleotides in the ocular tissue and their potential role in the pathogenesis of glaucoma. This article is part of the Special Issue on 'Purinergic Signaling: 50 years'.
Collapse
Affiliation(s)
- Youichi Shinozaki
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan; Interdisciplinary Brain-Immune Research Center, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Kozo Saito
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Kenji Kashiwagi
- Department of Ophthalmology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Schuichi Koizumi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan; Interdisciplinary Brain-Immune Research Center, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan.
| |
Collapse
|
6
|
Demais V, Pohl A, Wunderlich KA, Pfaller AM, Kaplan L, Barthélémy A, Dittrich R, Puig B, Giebel B, Hauck SM, Pfrieger FW, Grosche A. Release of VAMP5-positive extracellular vesicles by retinal Müller glia in vivo. J Extracell Vesicles 2022; 11:e12254. [PMID: 36043482 PMCID: PMC9428896 DOI: 10.1002/jev2.12254] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 06/25/2022] [Accepted: 07/18/2022] [Indexed: 11/11/2022] Open
Abstract
Cell-cell interactions in the central nervous system are based on the release of molecules mediating signal exchange and providing structural and trophic support through vesicular exocytosis and the formation of extracellular vesicles. The specific mechanisms employed by each cell type in the brain are incompletely understood. Here, we explored the means of communication used by Müller cells, a type of radial glial cells in the retina, which forms part of the central nervous system. Using immunohistochemical, electron microscopic, and molecular analyses, we provide evidence for the release of distinct extracellular vesicles from endfeet and microvilli of retinal Müller cells in adult mice in vivo. We identify VAMP5 as a Müller cell-specific SNARE component that is part of extracellular vesicles and responsive to ischemia, and we reveal differences between the secretomes of immunoaffinity-purified Müller cells and neurons in vitro. Our findings suggest extracellular vesicle-based communication as an important mediator of cellular interactions in the retina.
Collapse
Affiliation(s)
- Valerie Demais
- Plateforme Imagerie In Vitro, CNRS UAR 3156, NeuropôleUniversity of StrasbourgStrasbourgFrance
| | - Anne Pohl
- Department of Physiological GenomicsBioMedical Center BMCLudwig‐Maximilian UniversityPlanegg‐MartinsriedGermany
- Institute of Human GeneticsUniversity of RegensburgRegensburgGermany
| | - Kirsten A. Wunderlich
- Department of Physiological GenomicsBioMedical Center BMCLudwig‐Maximilian UniversityPlanegg‐MartinsriedGermany
| | - Anna M. Pfaller
- Department of Physiological GenomicsBioMedical Center BMCLudwig‐Maximilian UniversityPlanegg‐MartinsriedGermany
| | - Lew Kaplan
- Department of Physiological GenomicsBioMedical Center BMCLudwig‐Maximilian UniversityPlanegg‐MartinsriedGermany
| | - Amelie Barthélémy
- Centre National de la Recherche ScientifiqueUniversité de StrasbourgInstitut des Neurosciences Cellulaires et IntégrativesStrasbourgFrance
| | - Robin Dittrich
- Institute for Transfusion MedicineUniversity Hospital EssenUniversity of Duisburg‐EssenEssenGermany
| | - Berta Puig
- Neurology DepartmentExperimental Research in Stroke and Inflammation (ERSI)University Medical Center Hamburg‐EppendorfHamburgGermany
| | - Bernd Giebel
- Institute for Transfusion MedicineUniversity Hospital EssenUniversity of Duisburg‐EssenEssenGermany
| | - Stefanie M. Hauck
- Metabolomics and Proteomics Core and Research Unit Protein ScienceHelmholtz‐Zentrum MünchenMünchenGermany
| | - Frank W. Pfrieger
- Plateforme Imagerie In Vitro, CNRS UAR 3156, NeuropôleUniversity of StrasbourgStrasbourgFrance
- Centre National de la Recherche ScientifiqueUniversité de StrasbourgInstitut des Neurosciences Cellulaires et IntégrativesStrasbourgFrance
| | - Antje Grosche
- Department of Physiological GenomicsBioMedical Center BMCLudwig‐Maximilian UniversityPlanegg‐MartinsriedGermany
| |
Collapse
|
7
|
Markitantova YV, Simirskii VN. The Role of the Purinergic Signaling System in the Control of Histogenesis, Homeostasis, and Pathogenesis of the Vertebrate Retina. Russ J Dev Biol 2021. [DOI: 10.1134/s1062360421060084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
8
|
Li X, Lv J, Li J, Ren X. Kir4.1 may represent a novel therapeutic target for diabetic retinopathy (Review). Exp Ther Med 2021; 22:1021. [PMID: 34373707 PMCID: PMC8343704 DOI: 10.3892/etm.2021.10453] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 05/28/2021] [Indexed: 12/27/2022] Open
Abstract
As the major cause of irreversible loss of vision in adults, diabetic retinopathy (DR) is one of the most serious complications of diabetes. The imbalance of the retinal microenvironment and destruction of the blood-retinal barrier have a significant role in the progression of DR. Inward rectifying potassium channel 4.1 (Kir4.1) is located on Müller cells and is closely related to potassium homeostasis, water balance and glutamate clearance in the whole retina. The present review discusses the functions of Kir4.1 in regulating the retinal microenvironment and related biological mechanisms in DR. In the future, Kir4.1 may represent a novel alternative therapeutic target for DR through affecting the retinal microenvironment.
Collapse
Affiliation(s)
- Xiaoyu Li
- Department of Histology and Embryology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China.,Department of Radiotherapy Oncology, The Second Hospital of Dalian Medical University, Dalian, Liaoning 116023, P.R. China
| | - Jiajun Lv
- Department of Histology and Embryology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China.,Department of Radiotherapy Oncology, The Second Hospital of Dalian Medical University, Dalian, Liaoning 116023, P.R. China
| | - Jiazhi Li
- Department of Radiotherapy Oncology, The Second Hospital of Dalian Medical University, Dalian, Liaoning 116023, P.R. China
| | - Xiang Ren
- Department of Histology and Embryology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| |
Collapse
|
9
|
Harsing LG, Szénási G, Zelles T, Köles L. Purinergic-Glycinergic Interaction in Neurodegenerative and Neuroinflammatory Disorders of the Retina. Int J Mol Sci 2021; 22:ijms22126209. [PMID: 34201404 PMCID: PMC8228622 DOI: 10.3390/ijms22126209] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/03/2021] [Accepted: 06/04/2021] [Indexed: 12/26/2022] Open
Abstract
Neurodegenerative–neuroinflammatory disorders of the retina seriously hamper human vision. In searching for key factors that contribute to the development of these pathologies, we considered potential interactions among purinergic neuromodulation, glycinergic neurotransmission, and microglia activity in the retina. Energy deprivation at cellular levels is mainly due to impaired blood circulation leading to increased release of ATP and adenosine as well as glutamate and glycine. Interactions between these modulators and neurotransmitters are manifold. First, P2Y purinoceptor agonists facilitate reuptake of glycine by glycine transporter 1, while its inhibitors reduce reverse-mode operation; these events may lower extracellular glycine levels. The consequential changes in extracellular glycine concentration can lead to parallel changes in the activity of NR1/NR2B type NMDA receptors of which glycine is a mandatory agonist, and thereby may reduce neurodegenerative events in the retina. Second, P2Y purinoceptor agonists and glycine transporter 1 inhibitors may indirectly inhibit microglia activity by decreasing neuronal or glial glycine release in energy-compromised retina. These inhibitions may have a role in microglia activation, which is present during development and progression of neurodegenerative disorders such as glaucomatous and diabetic retinopathies and age-related macular degeneration or loss of retinal neurons caused by thromboembolic events. We have hypothesized that glycine transporter 1 inhibitors and P2Y purinoceptor agonists may have therapeutic importance in neurodegenerative–neuroinflammatory disorders of the retina by decreasing NR1/NR2B NMDA receptor activity and production and release of a series of proinflammatory cytokines from microglial cells.
Collapse
Affiliation(s)
- Laszlo G. Harsing
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary; (T.Z.); (L.K.)
- Correspondence: ; Tel.: +36-1-210-4416
| | - Gábor Szénási
- Institute of Translational Medicine, Semmelweis University, H-1089 Budapest, Hungary;
| | - Tibor Zelles
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary; (T.Z.); (L.K.)
- Department of Oral Biology, Semmelweis University, H-1089 Budapest, Hungary
| | - László Köles
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary; (T.Z.); (L.K.)
- Department of Oral Biology, Semmelweis University, H-1089 Budapest, Hungary
| |
Collapse
|
10
|
Díaz-Lezama N, Wolf A, Koch S, Pfaller AM, Biber J, Guillonneau X, Langmann T, Grosche A. PDGF Receptor Alpha Signaling Is Key for Müller Cell Homeostasis Functions. Int J Mol Sci 2021; 22:ijms22031174. [PMID: 33503976 PMCID: PMC7865899 DOI: 10.3390/ijms22031174] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/21/2021] [Accepted: 01/21/2021] [Indexed: 01/09/2023] Open
Abstract
Müller cells, the major retinal macroglia, are key to maintaining vascular integrity as well as retinal fluid and ion homeostasis. Although platelet derived growth factor (PDGF) receptor expression in Müller glia has been reported earlier, their actual role for Müller cell function and intimate interaction with cells of the retinal neurovascular unit remains unclear. To close this gap of knowledge, Müller cell-specific PDGF receptor alpha (PDGFRα) knockout (KO) mice were generated, characterized, and subjected to a model of choroidal neovascularization (CNV). PDGFRα-deficient Müller cells could not counterbalance hypoosmotic stress as efficiently as their wildtype counterparts. In wildtypes, the PDGFRα ligand PDGF-BB prevented Müller cell swelling induced by the administration of barium ions. This effect could be blocked by the PDGFR family inhibitor AC710. PDGF-BB could not restore the capability of an efficient volume regulation in PDGFRα KO Müller cells. Additionally, PDGFRα KO mice displayed reduced rod and cone-driven light responses. Altogether, these findings suggest that Müller glial PDGFRα is central for retinal functions under physiological conditions. In contrast, Müller cell-specific PDGFRα KO resulted in less vascular leakage and smaller lesion areas in the CNV model. Of note, the effect size was comparable to pharmacological blockade of PDGF signaling alone or in combination with anti-vascular endothelial growth factor (VEGF) therapy—a treatment regimen currently being tested in clinical trials. These data imply that targeting PDGF to treat retinal neovascular diseases may have short-term beneficial effects, but may elicit unwarranted side effects given the putative negative effects on Müller cell homeostatic functions potentially interfering with a long-term positive outcome.
Collapse
Affiliation(s)
- Nundehui Díaz-Lezama
- Department of Physiological Genomics, Biomedical Center, Ludwig-Maximilians-Universität München, D-82152 Planegg-Martinsried, Germany; (N.D.-L.); (S.K.); (A.M.P.); (J.B.)
| | - Anne Wolf
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50931 Cologne, Germany; (A.W.); (T.L.)
| | - Susanne Koch
- Department of Physiological Genomics, Biomedical Center, Ludwig-Maximilians-Universität München, D-82152 Planegg-Martinsried, Germany; (N.D.-L.); (S.K.); (A.M.P.); (J.B.)
| | - Anna M. Pfaller
- Department of Physiological Genomics, Biomedical Center, Ludwig-Maximilians-Universität München, D-82152 Planegg-Martinsried, Germany; (N.D.-L.); (S.K.); (A.M.P.); (J.B.)
| | - Josef Biber
- Department of Physiological Genomics, Biomedical Center, Ludwig-Maximilians-Universität München, D-82152 Planegg-Martinsried, Germany; (N.D.-L.); (S.K.); (A.M.P.); (J.B.)
| | - Xavier Guillonneau
- Institut de la Vision, INSERM, CNRS, Sorbonne Université, F-75012 Paris, France;
| | - Thomas Langmann
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50931 Cologne, Germany; (A.W.); (T.L.)
| | - Antje Grosche
- Department of Physiological Genomics, Biomedical Center, Ludwig-Maximilians-Universität München, D-82152 Planegg-Martinsried, Germany; (N.D.-L.); (S.K.); (A.M.P.); (J.B.)
- Correspondence:
| |
Collapse
|
11
|
Pauly D, Agarwal D, Dana N, Schäfer N, Biber J, Wunderlich KA, Jabri Y, Straub T, Zhang NR, Gautam AK, Weber BHF, Hauck SM, Kim M, Curcio CA, Stambolian D, Li M, Grosche A. Cell-Type-Specific Complement Expression in the Healthy and Diseased Retina. Cell Rep 2019; 29:2835-2848.e4. [PMID: 31775049 PMCID: PMC6911814 DOI: 10.1016/j.celrep.2019.10.084] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 05/24/2019] [Accepted: 10/22/2019] [Indexed: 02/06/2023] Open
Abstract
Complement dysregulation is a feature of many retinal diseases, yet mechanistic understanding at the cellular level is limited. Given this knowledge gap about which retinal cells express complement, we performed single-cell RNA sequencing on ~92,000 mouse retinal cells and validated our results in five major purified retinal cell types. We found evidence for a distributed cell-type-specific complement expression across 11 cell types. Notably, Müller cells are the major contributor of complement activators c1s, c3, c4, and cfb. Retinal pigment epithelium (RPE) mainly expresses cfh and the terminal complement components, whereas cfi and cfp transcripts are most abundant in neurons. Aging enhances c1s, cfb, cfp, and cfi expression, while cfh expression decreases. Transient retinal ischemia increases complement expression in microglia, Müller cells, and RPE. In summary, we report a unique complement expression signature for murine retinal cell types suggesting a well-orchestrated regulation of local complement expression in the retinal microenvironment. Overshooting complement activity contributes to retinal degeneration. Pauly et al. demonstrate a distinct complement expression profile of retinal cell types that changes with aging and during retinal degeneration. This prompts the intriguing concept of a local retinal complement activation possibly independent of the systemic components typically produced by the liver.
Collapse
Affiliation(s)
- Diana Pauly
- Experimental Ophthalmology, University Hospital Regensburg, Regensburg 93053, Germany.
| | - Divyansh Agarwal
- Genomics and Computational Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nicholas Dana
- Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nicole Schäfer
- Experimental Ophthalmology, University Hospital Regensburg, Regensburg 93053, Germany
| | - Josef Biber
- Department of Physiological Genomics, Biomedical Center, Ludwig Maximilians University Munich, Planegg-Martinsried 82152, Germany
| | - Kirsten A Wunderlich
- Department of Physiological Genomics, Biomedical Center, Ludwig Maximilians University Munich, Planegg-Martinsried 82152, Germany
| | - Yassin Jabri
- Experimental Ophthalmology, University Hospital Regensburg, Regensburg 93053, Germany
| | - Tobias Straub
- Core Facility Bioinformatics, Biomedical Center, Ludwig Maximilians University Munich, Planegg-Martinsried 82152, Germany
| | - Nancy R Zhang
- Department of Statistics, The Wharton School, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Avneesh K Gautam
- Department of Medicine, Immunology and Allergy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Bernhard H F Weber
- Institute of Human Genetics, University of Regensburg, Regensburg 93053, Germany
| | - Stefanie M Hauck
- Research Unit Protein Science, Helmholtz Center Munich, Research Center for Environmental Health (GmbH), Munich 80939, Germany
| | - Mijin Kim
- Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Christine A Curcio
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham, AL 35294-0019, USA
| | - Dwight Stambolian
- Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mingyao Li
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Antje Grosche
- Department of Physiological Genomics, Biomedical Center, Ludwig Maximilians University Munich, Planegg-Martinsried 82152, Germany.
| |
Collapse
|
12
|
Mages K, Grassmann F, Jägle H, Rupprecht R, Weber BHF, Hauck SM, Grosche A. The agonistic TSPO ligand XBD173 attenuates the glial response thereby protecting inner retinal neurons in a murine model of retinal ischemia. J Neuroinflammation 2019; 16:43. [PMID: 30777091 PMCID: PMC6378755 DOI: 10.1186/s12974-019-1424-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Accepted: 01/31/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Ligand-driven modulation of the mitochondrial translocator protein 18 kDa (TSPO) was recently described to dampen the neuroinflammatory response of microglia in a retinal light damage model resulting in protective effects on photoreceptors. We characterized the effects of the TSPO ligand XBD173 in the postischemic retina focusing on changes in the response pattern of the major glial cell types of the retina-microglia and Müller cells. METHODS Retinal ischemia was induced by increasing the intraocular pressure for 60 min followed by reperfusion of the tissue in mice. On retinal cell types enriched via immunomagnetic separation expression analysis of TSPO, its ligand diazepam-binding inhibitor (DBI) and markers of glial activation were performed at transcript and protein level using RNA sequencing, qRT-PCR, lipid chromatography-mass spectrometry, and immunofluorescent labeling. Data on cell morphology and numbers were assessed in retinal slice and flatmount preparations. The retinal functional integrity was determined by electroretinogram recordings. RESULTS We demonstrate that TSPO is expressed by Müller cells, microglia, vascular cells, retinal pigment epithelium (RPE) of the healthy and postischemic retina, but only at low levels in retinal neurons. While an alleviated neurodegeneration upon XBD173 treatment was found in postischemic retinae as compared to vehicle controls, this neuroprotective effect of XBD173 is mediated putatively by its action on retinal glia. After transient ischemia, TSPO as a marker of activation was upregulated to similar levels in microglia as compared to their counterparts in healthy retinae irrespective of the treatment regimen. However, less microglia were found in XBD173-treated postischemic retinae at 3 days post-surgery (dps) which displayed a more ramified morphology than in retinae of vehicle-treated mice indicating a dampened microglia activation. Müller cells, the major retinal macroglia, show upregulation of the typical gliosis marker GFAP. Importantly, glutamine synthetase was more stably expressed in Müller glia of XBD173-treated postischemic retinae and homeostatic functions such as cellular volume regulation typically diminished in gliotic Müller cells remained functional. CONCLUSIONS In sum, our data imply that beneficial effects of XBD173 treatment on the postischemic survival of inner retinal neurons were primarily mediated by stabilizing neurosupportive functions of glial cells.
Collapse
Affiliation(s)
- Kristin Mages
- Institute of Human Genetics, University of Regensburg, Franz-Josef-Strauß-Allee 11, 93053, Regensburg, Germany
| | - Felix Grassmann
- Institute of Human Genetics, University of Regensburg, Franz-Josef-Strauß-Allee 11, 93053, Regensburg, Germany.,Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Nobels väg 12A, Stockholm, Sweden
| | - Herbert Jägle
- Department of Ophthalmology, University of Regensburg, Franz-Josef-Strauß-Allee 11, 93053, Regensburg, Germany
| | - Rainer Rupprecht
- Department of Psychiatry and Psychotherapy, University of Regensburg, Universitätsstraße 84, 93053, Regensburg, Germany
| | - Bernhard H F Weber
- Institute of Human Genetics, University of Regensburg, Franz-Josef-Strauß-Allee 11, 93053, Regensburg, Germany
| | - Stefanie M Hauck
- Research Unit Protein Science, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Heidemannstraße 1, 80939, Munich, Germany
| | - Antje Grosche
- Institute of Human Genetics, University of Regensburg, Franz-Josef-Strauß-Allee 11, 93053, Regensburg, Germany. .,Department of Physiological Genomics, Ludwig-Maximilians-Universität München, Großhaderner Str. 9, 82152, Planegg-Martinsried, Germany.
| |
Collapse
|
13
|
Kaczmarek-Hajek K, Zhang J, Kopp R, Grosche A, Rissiek B, Saul A, Bruzzone S, Engel T, Jooss T, Krautloher A, Schuster S, Magnus T, Stadelmann C, Sirko S, Koch-Nolte F, Eulenburg V, Nicke A. Re-evaluation of neuronal P2X7 expression using novel mouse models and a P2X7-specific nanobody. eLife 2018; 7:36217. [PMID: 30074479 PMCID: PMC6140716 DOI: 10.7554/elife.36217] [Citation(s) in RCA: 120] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 07/31/2018] [Indexed: 12/18/2022] Open
Abstract
The P2X7 channel is involved in the pathogenesis of various CNS diseases. An increasing number of studies suggest its presence in neurons where its putative functions remain controversial for more than a decade. To resolve this issue and to provide a model for analysis of P2X7 functions, we generated P2X7 BAC transgenic mice that allow visualization of functional EGFP-tagged P2X7 receptors in vivo. Extensive characterization of these mice revealed dominant P2X7-EGFP protein expression in microglia, Bergmann glia, and oligodendrocytes, but not in neurons. These findings were further validated by microglia- and oligodendrocyte-specific P2X7 deletion and a novel P2X7-specific nanobody. In addition to the first quantitative analysis of P2X7 protein expression in the CNS, we show potential consequences of its overexpression in ischemic retina and post-traumatic cerebral cortex grey matter. This novel mouse model overcomes previous limitations in P2X7 research and will help to determine its physiological roles and contribution to diseases. The human body relies on a molecule called ATP as an energy source and as a messenger. When cells die, for example if they are damaged or because of inflammation, they release large amounts of ATP into their environment. Their neighbors can detect the outpouring of ATP through specific receptors, the proteins that sit at the cell’s surface and can bind external agents. Scientists believe that one of these ATP-binding receptors, P2X7, responds to high levels of ATP by triggering a cascade of reactions that results in inflammation and cell death. P2X7 also seems to play a role in several brain diseases such as epilepsia and Alzheimer’s, but the exact mechanisms are not known. In particular, how this receptor is involved in the death of neurons is unclear, and researchers still debate whether P2X7 is present in neurons and in other types of brain cells. To answer this, Kaczmarek-Hájek, Zhang, Kopp et al. created genetically modified mice in which the P2X7 receptors carry a fluorescent dye. Powerful microscopes can pick up the light signal from the dye and help to reveal which cells have the receptors. These experiments show that neurons do not carry the protein; instead, P2X7 is present in certain brain cells that keep the neurons healthy. For example, it is found in the immune cells that ‘clean up’ the organ, and the cells that support and insulate neurons. Kaczmarek-Hájek et al. further provide preliminary data suggesting that, under certain conditions, if too many P2X7 receptors are present in these cells neuronal damage might be increased. It is therefore possible that the brain cells that carry P2X7 indirectly contribute to the death of neurons when large amounts of ATP are released. The genetically engineered mouse designed for the experiments could be used in further studies to dissect the role that P2X7 plays in diseases of the nervous system. In particular, this mouse model might help to understand whether the receptor could become a drug target for neurodegenerative conditions.
Collapse
Affiliation(s)
- Karina Kaczmarek-Hajek
- Department of Molecular Biology of Neuronal Signals, Max Planck Institute for Experimental Medicine, Göttingen, Germany
| | - Jiong Zhang
- Department of Molecular Biology of Neuronal Signals, Max Planck Institute for Experimental Medicine, Göttingen, Germany.,Walther Straub Institute for Pharmacology and Toxicology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Robin Kopp
- Walther Straub Institute for Pharmacology and Toxicology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Antje Grosche
- Institute for Human Genetics, University of Regensburg, Regensburg, Germany.,Department of Physiological Genomics, Ludwig-Maximilians-Universität München, München, Germany
| | - Björn Rissiek
- Department of Neurology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Anika Saul
- Department of Molecular Biology of Neuronal Signals, Max Planck Institute for Experimental Medicine, Göttingen, Germany
| | - Santina Bruzzone
- Department of Experimental Medicine and CEBR, University of Genova, Genova, Italy
| | - Tobias Engel
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Tina Jooss
- Walther Straub Institute for Pharmacology and Toxicology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Anna Krautloher
- Walther Straub Institute for Pharmacology and Toxicology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Stefanie Schuster
- Institute of Biochemistry, University Erlangen-Nürnberg, Erlangen, Germany
| | - Tim Magnus
- Department of Neurology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | | | - Swetlana Sirko
- Department of Physiological Genomics, Ludwig-Maximilians-Universität München, München, Germany.,Institute of Stem Cell Research, Helmholtz Center Munich, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Friedrich Koch-Nolte
- Department of Immunology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Volker Eulenburg
- Institute of Biochemistry, University Erlangen-Nürnberg, Erlangen, Germany.,Department of Anaesthesiology and Intensive Care Therapy, University of Leipzig, Leipzig, Germany
| | - Annette Nicke
- Department of Molecular Biology of Neuronal Signals, Max Planck Institute for Experimental Medicine, Göttingen, Germany.,Walther Straub Institute for Pharmacology and Toxicology, Ludwig-Maximilians-Universität München, Munich, Germany
| |
Collapse
|
14
|
Pannicke T, Wagner L, Reichenbach A, Grosche A. Electrophysiological characterization of Müller cells from the ischemic retina of mice deficient in the leukemia inhibitory factor. Neurosci Lett 2018; 670:69-74. [PMID: 29391217 DOI: 10.1016/j.neulet.2018.01.047] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 01/23/2018] [Accepted: 01/24/2018] [Indexed: 12/24/2022]
Abstract
Leukemia inhibitory factor (LIF) is a cytokine that exerts different effects in the nervous system. It is involved in neuronal injuries and diseases and is assumed to be neuroprotective and to regulate reactive gliosis. In LIF-deficient (LIF-/-) mice, expression of glial fibrillary acidic protein in retinal Müller glial cells as a hallmark of reactive gliosis is suppressed during retinal degenerations. Here, we detected expression of LIF and its receptors in Müller cells of the murine retina. Moreover, electrophysiological alterations of Müller cells 7 days after transient retinal ischemia were studied by the patch-clamp technique. The amplitude of inward currents in Müller cells from the postischemic retina was reduced to 51% in wild type and to 70% in LIF-/- mice. This demonstrates that decrease of inward currents takes place in reactive Müller cells even in the absence of LIF.
Collapse
Affiliation(s)
- Thomas Pannicke
- Paul-Flechsig-Institut für Hirnforschung, Universität Leipzig, Liebigstr. 19, D-04103 Leipzig, Germany.
| | - Lysann Wagner
- Paul-Flechsig-Institut für Hirnforschung, Universität Leipzig, Liebigstr. 19, D-04103 Leipzig, Germany
| | - Andreas Reichenbach
- Paul-Flechsig-Institut für Hirnforschung, Universität Leipzig, Liebigstr. 19, D-04103 Leipzig, Germany
| | - Antje Grosche
- Department of Physiological Genomics, Ludwig-Maximilians-Universität München, Großhaderner Str. 9, D-82152 Planegg-Martinsried, Germany
| |
Collapse
|
15
|
Abstract
Müller glia, the principal macroglia of the retina, express diverse subtypes of adenosine and metabotropic purinergic (P2Y) receptors. Müller cells of several species, including man, also express ionotropic P2X7 receptors. ATP is liberated from Müller cells after activation of metabotropic glutamate receptors and during osmotic and mechanical induction of membrane stretch; adenosine is released through equilibrative nucleoside transporters. Müller cell-derived purines modulate the neuronal activity and have autocrine effects, for example, induction of glial calcium waves and regulation of the cellular volume. Glial calcium waves induced by neuron-derived ATP mediate functional hyperemia in the retina. Purinergic signaling contributes to the induction of Müller cell gliosis, for example, of cellular proliferation and downregulation of potassium channels, which are important for the homeostatic functions of Müller cells. Purinergic glial calcium waves may also promote the long-range propagation of gliosis and neuronal degeneration across the retinal tissue. The osmotic ATP release is inhibited under pathological conditions. Inhibition of the ATP release may result in osmotic Müller cell swelling and dysregulation of the water transport through the cells; both may contribute to the development of retinal edema. Suppression of the osmotic ATP release and upregulation of the ecto-apyrase (NTPDase1), which facilitate the extracellular degradation of ATP and the formation of adenosine, may protect neurons and photoreceptors from death due to overactivation of P2X receptors. Pharmacological inhibition of P2X7 receptors and stimulation of adenosine receptors may represent clinical approaches to prevent retinal cell death and dysregulated cell proliferation, and to treat retinal edema.
Collapse
Affiliation(s)
- Andreas Reichenbach
- 1 Paul Flechsig Institute of Brain Research, University of Leipzig , Leipzig, Germany
| | - Andreas Bringmann
- 2 Department of Ophthalmology and Eye Hospital, University of Leipzig , Leipzig, Germany
| |
Collapse
|
16
|
Lu Q, Livi GP, Modha S, Yusa K, Macarrón R, Dow DJ. Applications of CRISPR genome editing technology in drug target identification and validation. Expert Opin Drug Discov 2017; 12:541-552. [DOI: 10.1080/17460441.2017.1317244] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
17
|
Wagner L, Pannicke T, Rupprecht V, Frommherz I, Volz C, Illes P, Hirrlinger J, Jägle H, Egger V, Haydon PG, Pfrieger FW, Grosche A. Suppression of SNARE-dependent exocytosis in retinal glial cells and its effect on ischemia-induced neurodegeneration. Glia 2017; 65:1059-1071. [PMID: 28370368 PMCID: PMC5485027 DOI: 10.1002/glia.23144] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 03/03/2017] [Accepted: 03/13/2017] [Indexed: 12/25/2022]
Abstract
Nervous tissue is characterized by a tight structural association between glial cells and neurons. It is well known that glial cells support neuronal functions, but their role under pathologic conditions is less well understood. Here, we addressed this question in vivo using an experimental model of retinal ischemia and transgenic mice for glia‐specific inhibition of soluble N‐ethylmaleimide‐sensitive factor attachment protein receptor (SNARE)‐dependent exocytosis. Transgene expression reduced glutamate, but not ATP release from single Müller cells, impaired glial volume regulation under normal conditions and reduced neuronal dysfunction and death in the inner retina during the early stages of ischemia. Our study reveals that the SNARE‐dependent exocytosis in glial cells contributes to neurotoxicity during ischemia in vivo and suggests glial exocytosis as a target for therapeutic approaches.
Collapse
Affiliation(s)
- Lysann Wagner
- Paul Flechsig Institute of Brain Research, University of Leipzig, Liebigstr. 19, Leipzig, 04103, Germany
| | - Thomas Pannicke
- Paul Flechsig Institute of Brain Research, University of Leipzig, Liebigstr. 19, Leipzig, 04103, Germany
| | - Vanessa Rupprecht
- Institute of Zoology, University of Regensburg, Universitätsstr. 31, Regensburg, 93040, Germany
| | - Ina Frommherz
- Paul Flechsig Institute of Brain Research, University of Leipzig, Liebigstr. 19, Leipzig, 04103, Germany
| | - Cornelia Volz
- Department of Ophthalmology, University of Regensburg, Franz-Josef-Strauß-Allee 1, Regensburg, 93953, Germany
| | - Peter Illes
- Rudolf Boehm Institute for Pharmacology and Toxicology, University of Leipzig, Härtelstr. 16/18, 04107, Leipzig, Germany
| | - Johannes Hirrlinger
- Carl Ludwig Institute of Physiology, University of Leipzig, Liebigstr. 27, Leipzig, 04103, Germany.,Department of Neurogenetics, Max-Planck-Institute for Experimental Medicine, Hermann-Rein-Str. 3, Göttingen, 37075, Germany
| | - Herbert Jägle
- Department of Ophthalmology, University of Regensburg, Franz-Josef-Strauß-Allee 1, Regensburg, 93953, Germany
| | - Veronica Egger
- Institute of Zoology, University of Regensburg, Universitätsstr. 31, Regensburg, 93040, Germany
| | - Philip G Haydon
- Department of Neuroscience, Tufts University School of Medicine, 136 Harrison Avenue, Boston, Massachusetts, 02111, USA
| | - Frank W Pfrieger
- Institute of Cellular and Integrative Neurosciences, CNRS UPR 3212, University of Strasbourg, 5 rue Blaise Pascal, Strasbourg Cedex, 67084, France
| | - Antje Grosche
- Institute of Human Genetics, University of Regensburg, Franz-Josef-Strauß-Allee 1, Regensburg, 93953, Germany
| |
Collapse
|
18
|
Jacobson KA, Civan MM. Ocular Purine Receptors as Drug Targets in the Eye. J Ocul Pharmacol Ther 2016; 32:534-547. [PMID: 27574786 PMCID: PMC5069731 DOI: 10.1089/jop.2016.0090] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 07/13/2016] [Indexed: 02/06/2023] Open
Abstract
Agonists and antagonists of various subtypes of G protein coupled adenosine receptors (ARs), P2Y receptors (P2YRs), and ATP-gated P2X receptor ion channels (P2XRs) are under consideration as agents for the treatment of ocular diseases, including glaucoma and dry eye. Numerous nucleoside and nonnucleoside modulators of the receptors are available as research tools and potential therapeutic molecules. Three of the 4 subtypes of ARs have been exploited with clinical candidate molecules for treatment of the eye: A1, A2A, and A3. An A1AR agonist is in clinical trials for glaucoma, A2AAR reduces neuroinflammation, A3AR protects retinal ganglion cells from apoptosis, and both A3AR agonists and antagonists had been reported to lower intraocular pressure (IOP). Extracellular concentrations of endogenous nucleotides, including dinucleoside polyphosphates, are increased in pathological states, activating P2Y and P2XRs throughout the eye. P2YR agonists, including P2Y2 and P2Y6, lower IOP. Antagonists of the P2X7R prevent the ATP-induced neuronal apoptosis in the retina. Thus, modulators of the purinome in the eye might be a source of new therapies for ocular diseases.
Collapse
Affiliation(s)
- Kenneth A. Jacobson
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Mortimer M. Civan
- Departments of Physiology and Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
19
|
Effects of IP3R2 Receptor Deletion in the Ischemic Mouse Retina. Neurochem Res 2015; 41:677-86. [PMID: 26446037 DOI: 10.1007/s11064-015-1735-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 09/24/2015] [Accepted: 09/30/2015] [Indexed: 10/23/2022]
Abstract
Glial cells in the diseased nervous system undergo a process known as reactive gliosis. Gliosis of retinal Müller glial cells is characterized by an upregulation of glial fibrillary acidic protein and frequently by a reduction of inward K(+) current amplitudes. Purinergic signaling is assumed to be involved in gliotic processes. As previously shown, lack of the nucleotide receptor P2Y1 leads to an altered regulation of K(+) currents in Müller cells of the ischemic retina. Here, we asked first whether this effect is mediated by the IP3 receptor subtype 2 (IP3R2) known as the major downstream signaling target of P2Y1 in Müller cells. The second question was whether lack of IP3R2 affects neuronal survival in the control and ischemic retina. Ischemia was induced in wild type and IP3R2-deficient (IP 3 R2 (-/-)) mice by transient elevation of the intraocular pressure. Immunostaining and TUNEL labelling were used to quantify neuronal cell loss. The downregulation of inward K(+) currents in Müller cells from ischemic IP 3 R2 (-/-) retinae was less strong than in wild type animals. The reduction of the number of cells in the ganglion cell layer and of calretinin- and calbindin-positive cells 7 days after ischemia was similar in wild type and IP 3 R2 (-/-) mice. However, IP3R2 deficiency led to an increased number of TUNEL-positive cells in the outer nuclear layer at 1 day and to an enhanced postischemic loss of photoreceptors 7 days after ischemia. This implies that IP3R2 is involved in some but not all aspects of signaling in Müller cells after an ischemic insult.
Collapse
|
20
|
Grosche A, Hauser A, Lepper MF, Mayo R, von Toerne C, Merl-Pham J, Hauck SM. The Proteome of Native Adult Müller Glial Cells From Murine Retina. Mol Cell Proteomics 2015; 15:462-80. [PMID: 26324419 PMCID: PMC4739667 DOI: 10.1074/mcp.m115.052183] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Indexed: 12/26/2022] Open
Abstract
To date, the proteomic profiling of Müller cells, the dominant macroglia of the retina, has been hampered because of the absence of suitable enrichment methods. We established a novel protocol to isolate native, intact Müller cells from adult murine retinae at excellent purity which retain in situ morphology and are well suited for proteomic analyses. Two different strategies of sample preparation - an in StageTips (iST) and a subcellular fractionation approach including cell surface protein profiling were used for quantitative liquid chromatography-mass spectrometry (LC-MSMS) comparing Müller cell-enriched to depleted neuronal fractions. Pathway enrichment analyses on both data sets enabled us to identify Müller cell-specific functions which included focal adhesion kinase signaling, signal transduction mediated by calcium as second messenger, transmembrane neurotransmitter transport and antioxidant activity. Pathways associated with RNA processing, cellular respiration and phototransduction were enriched in the neuronal subpopulation. Proteomic results were validated for selected Müller cell genes by quantitative real time PCR, confirming the high expression levels of numerous members of the angiogenic and anti-inflammatory annexins and antioxidant enzymes (e.g. paraoxonase 2, peroxiredoxin 1, 4 and 6). Finally, the significant enrichment of antioxidant proteins in Müller cells was confirmed by measurements on vital retinal cells using the oxidative stress indicator CM-H2DCFDA. In contrast to photoreceptors or bipolar cells, Müller cells were most efficiently protected against H2O2-induced reactive oxygen species formation, which is in line with the protein repertoire identified in the proteomic profiling. Our novel approach to isolate intact glial cells from adult retina in combination with proteomic profiling enabled the identification of novel Müller glia specific proteins, which were validated as markers and for their functional impact in glial physiology. This provides the basis to allow the discovery of novel glial specializations and will enable us to elucidate the role of Müller cells in retinal pathologies — a topic still controversially discussed.
Collapse
Affiliation(s)
- Antje Grosche
- From the ‡Insitute of Human Genetics, University of Regensburg, D-93053 Regensburg, Germany;
| | - Alexandra Hauser
- From the ‡Insitute of Human Genetics, University of Regensburg, D-93053 Regensburg, Germany
| | - Marlen Franziska Lepper
- §Research Unit Protein Science, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), D-85764 Neuherberg, Germany
| | - Rebecca Mayo
- From the ‡Insitute of Human Genetics, University of Regensburg, D-93053 Regensburg, Germany
| | - Christine von Toerne
- §Research Unit Protein Science, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), D-85764 Neuherberg, Germany
| | - Juliane Merl-Pham
- §Research Unit Protein Science, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), D-85764 Neuherberg, Germany
| | - Stefanie M Hauck
- §Research Unit Protein Science, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), D-85764 Neuherberg, Germany
| |
Collapse
|
21
|
Wunderlich KA, Tanimoto N, Grosche A, Zrenner E, Pekny M, Reichenbach A, Seeliger MW, Pannicke T, Perez MT. Retinal functional alterations in mice lacking intermediate filament proteins glial fibrillary acidic protein and vimentin. FASEB J 2015; 29:4815-28. [PMID: 26251181 DOI: 10.1096/fj.15-272963] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 07/27/2015] [Indexed: 01/02/2023]
Abstract
Vimentin (Vim) and glial fibrillary acidic protein (GFAP) are important components of the intermediate filament (IF) (or nanofilament) system of astroglial cells. We conducted full-field electroretinogram (ERG) recordings and found that whereas photoreceptor responses (a-wave) were normal in uninjured GFAP(-/-)Vim(-/-) mice, b-wave amplitudes were increased. Moreover, we found that Kir (inward rectifier K(+)) channel protein expression was reduced in the retinas of GFAP(-/-)Vim(-/-) mice and that Kir-mediated current amplitudes were lower in Müller glial cells isolated from these mice. Studies have shown that the IF system, in addition, is involved in the retinal response to injury and that attenuated Müller cell reactivity and reduced photoreceptor cell loss are observed in IF-deficient mice after experimental retinal detachment. We investigated whether the lack of IF proteins would affect cell survival in a retinal ischemia-reperfusion model. We found that although cell loss was induced in both genotypes, the number of surviving cells in the inner retina was lower in IF-deficient mice. Our findings thus show that the inability to produce GFAP and Vim affects normal retinal physiology and that the effect of IF deficiency on retinal cell survival differs, depending on the underlying pathologic condition.
Collapse
Affiliation(s)
- Kirsten A Wunderlich
- *Department of Clinical Sciences, Division of Ophthalmology, and NanoLund, Nanometer Structure Consortium, Lund University, Lund, Sweden; Graduate School of Cellular and Molecular Neuroscience, Center for Integrative Neuroscience (CIN), and Division of Ocular Neurodegeneration, Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany; Institute of Human Genetics, University of Regensburg, Regensburg, Germany; Center for Brain Repair and Rehabilitation, Department of Clinical Neuroscience and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia; **Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia; and Paul Flechsig Institute of Brain Research, University of Leipzig, Leipzig, Germany
| | - Naoyuki Tanimoto
- *Department of Clinical Sciences, Division of Ophthalmology, and NanoLund, Nanometer Structure Consortium, Lund University, Lund, Sweden; Graduate School of Cellular and Molecular Neuroscience, Center for Integrative Neuroscience (CIN), and Division of Ocular Neurodegeneration, Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany; Institute of Human Genetics, University of Regensburg, Regensburg, Germany; Center for Brain Repair and Rehabilitation, Department of Clinical Neuroscience and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia; **Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia; and Paul Flechsig Institute of Brain Research, University of Leipzig, Leipzig, Germany
| | - Antje Grosche
- *Department of Clinical Sciences, Division of Ophthalmology, and NanoLund, Nanometer Structure Consortium, Lund University, Lund, Sweden; Graduate School of Cellular and Molecular Neuroscience, Center for Integrative Neuroscience (CIN), and Division of Ocular Neurodegeneration, Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany; Institute of Human Genetics, University of Regensburg, Regensburg, Germany; Center for Brain Repair and Rehabilitation, Department of Clinical Neuroscience and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia; **Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia; and Paul Flechsig Institute of Brain Research, University of Leipzig, Leipzig, Germany
| | - Eberhart Zrenner
- *Department of Clinical Sciences, Division of Ophthalmology, and NanoLund, Nanometer Structure Consortium, Lund University, Lund, Sweden; Graduate School of Cellular and Molecular Neuroscience, Center for Integrative Neuroscience (CIN), and Division of Ocular Neurodegeneration, Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany; Institute of Human Genetics, University of Regensburg, Regensburg, Germany; Center for Brain Repair and Rehabilitation, Department of Clinical Neuroscience and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia; **Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia; and Paul Flechsig Institute of Brain Research, University of Leipzig, Leipzig, Germany
| | - Milos Pekny
- *Department of Clinical Sciences, Division of Ophthalmology, and NanoLund, Nanometer Structure Consortium, Lund University, Lund, Sweden; Graduate School of Cellular and Molecular Neuroscience, Center for Integrative Neuroscience (CIN), and Division of Ocular Neurodegeneration, Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany; Institute of Human Genetics, University of Regensburg, Regensburg, Germany; Center for Brain Repair and Rehabilitation, Department of Clinical Neuroscience and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia; **Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia; and Paul Flechsig Institute of Brain Research, University of Leipzig, Leipzig, Germany
| | - Andreas Reichenbach
- *Department of Clinical Sciences, Division of Ophthalmology, and NanoLund, Nanometer Structure Consortium, Lund University, Lund, Sweden; Graduate School of Cellular and Molecular Neuroscience, Center for Integrative Neuroscience (CIN), and Division of Ocular Neurodegeneration, Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany; Institute of Human Genetics, University of Regensburg, Regensburg, Germany; Center for Brain Repair and Rehabilitation, Department of Clinical Neuroscience and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia; **Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia; and Paul Flechsig Institute of Brain Research, University of Leipzig, Leipzig, Germany
| | - Mathias W Seeliger
- *Department of Clinical Sciences, Division of Ophthalmology, and NanoLund, Nanometer Structure Consortium, Lund University, Lund, Sweden; Graduate School of Cellular and Molecular Neuroscience, Center for Integrative Neuroscience (CIN), and Division of Ocular Neurodegeneration, Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany; Institute of Human Genetics, University of Regensburg, Regensburg, Germany; Center for Brain Repair and Rehabilitation, Department of Clinical Neuroscience and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia; **Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia; and Paul Flechsig Institute of Brain Research, University of Leipzig, Leipzig, Germany
| | - Thomas Pannicke
- *Department of Clinical Sciences, Division of Ophthalmology, and NanoLund, Nanometer Structure Consortium, Lund University, Lund, Sweden; Graduate School of Cellular and Molecular Neuroscience, Center for Integrative Neuroscience (CIN), and Division of Ocular Neurodegeneration, Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany; Institute of Human Genetics, University of Regensburg, Regensburg, Germany; Center for Brain Repair and Rehabilitation, Department of Clinical Neuroscience and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia; **Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia; and Paul Flechsig Institute of Brain Research, University of Leipzig, Leipzig, Germany
| | - Maria-Thereza Perez
- *Department of Clinical Sciences, Division of Ophthalmology, and NanoLund, Nanometer Structure Consortium, Lund University, Lund, Sweden; Graduate School of Cellular and Molecular Neuroscience, Center for Integrative Neuroscience (CIN), and Division of Ocular Neurodegeneration, Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany; Institute of Human Genetics, University of Regensburg, Regensburg, Germany; Center for Brain Repair and Rehabilitation, Department of Clinical Neuroscience and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia; **Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia; and Paul Flechsig Institute of Brain Research, University of Leipzig, Leipzig, Germany
| |
Collapse
|
22
|
Reichenbach A, Bringmann A. Purinergic signaling in retinal degeneration and regeneration. Neuropharmacology 2015; 104:194-211. [PMID: 25998275 DOI: 10.1016/j.neuropharm.2015.05.005] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 05/07/2015] [Accepted: 05/07/2015] [Indexed: 02/01/2023]
Abstract
Purinergic signaling is centrally involved in mediating the degeneration of the injured and diseased retina, the induction of retinal gliosis, and the protection of the retinal tissue from degeneration. Dysregulated calcium signaling triggered by overactivation of P2X7 receptors is a crucial step in the induction of neuronal and microvascular cell death under pathogenic conditions like ischemia-hypoxia, elevated intraocular pressure, and diabetes, respectively. Overactivation of P2X7 plays also a pathogenic role in inherited and age-related photoreceptor cell death and in the age-related dysfunction and degeneration of the retinal pigment epithelium. Gliosis of micro- and macroglial cells, which is induced and/or modulated by purinergic signaling and associated with an impaired homeostatic support to neurons, and the ATP-mediated propagation of retinal gliosis from a focal injury into the surrounding noninjured tissue are involved in inducing secondary cell death in the retina. On the other hand, alterations in the glial metabolism of extracellular nucleotides, resulting in a decreased level of ATP and an increased level of adenosine, may be neuroprotective in the diseased retina. Purinergic signals stimulate the proliferation of retinal glial cells which contributes to glial scarring which has protective effects on retinal degeneration and adverse effects on retinal regeneration. Pharmacological modulation of purinergic receptors, e.g., inhibition of P2X and activation of adenosine receptors, may have clinical importance for the prevention of photoreceptor, neuronal, and microvascular cell death in diabetic retinopathy, retinitis pigmentosa, age-related macular degeneration, and glaucoma, respectively, for the clearance of retinal edema, and the inhibition of dysregulated cell proliferation in proliferative retinopathies. This article is part of a Special Issue entitled 'Purines in Neurodegeneration and Neuroregeneration'.
Collapse
Affiliation(s)
- Andreas Reichenbach
- Paul Flechsig Institute of Brain Research, University of Leipzig, Leipzig, Germany.
| | - Andreas Bringmann
- Department of Ophthalmology and Eye Hospital, University of Leipzig, Leipzig, Germany
| |
Collapse
|
23
|
|