1
|
Yang Z, Tian D, Zhao X, Luo Y, Chen Y. The gut-retina axis: Uncovering the role of autoimmunity in glaucoma development. Heliyon 2024; 10:e35516. [PMID: 39170439 PMCID: PMC11336731 DOI: 10.1016/j.heliyon.2024.e35516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 07/27/2024] [Accepted: 07/30/2024] [Indexed: 08/23/2024] Open
Abstract
Glaucoma, a leading cause of irreversible blindness worldwide, is characterized by progressive loss of retinal ganglion cells (RGCs) and optic nerve damage. While elevated intraocular pressure (IOP) is the only known modifiable risk factor, normal-tension glaucoma (NTG) challenges this notion, suggesting other mechanisms beyond IOP may contribute to its development. Emerging evidence support the hypothesis that glaucoma may be an autoimmune disease. This review summarizes evidence for this hypothesis, focusing on the gut-retina axis. We discuss how antigens of gut bacterial prime peripheral T cells to breach the blood-retina barrier (BRB) and initiate cross-reactivity with ocular tissues via molecular mimicry, resulting in autoimmune RGC damage. Understanding these mechanisms may uncover new diagnostic biomarkers and therapeutic strategies targeting immune pathways alongside conventional IOP-lowering treatments.
Collapse
Affiliation(s)
- Zuyi Yang
- Eight-year Medical Doctor Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dianzhe Tian
- Eight-year Medical Doctor Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xinyu Zhao
- Department of Ophthalmology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, 100730, China
- Key Lab of Ocular Fundus Diseases, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Yunping Luo
- Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
- Collaborative Innovation Center for Biotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Youxin Chen
- Department of Ophthalmology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, 100730, China
- Key Lab of Ocular Fundus Diseases, Chinese Academy of Medical Sciences, Beijing, 100730, China
| |
Collapse
|
2
|
Tsai T, Reinehr S, Deppe L, Strubbe A, Kluge N, Dick HB, Joachim SC. Glaucoma Animal Models beyond Chronic IOP Increase. Int J Mol Sci 2024; 25:906. [PMID: 38255979 PMCID: PMC10815097 DOI: 10.3390/ijms25020906] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 01/04/2024] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
Glaucoma is a complex and multifactorial disease defined as the loss of retinal ganglion cells (RGCs) and their axons. Besides an elevated intraocular pressure (IOP), other mechanisms play a pivotal role in glaucoma onset and progression. For example, it is known that excitotoxicity, immunological alterations, ischemia, and oxidative stress contribute to the neurodegeneration in glaucoma disease. To study these effects and to discover novel therapeutic approaches, appropriate animal models are needed. In this review, we focus on various glaucoma animal models beyond an elevated IOP. We introduce genetically modified mice, e.g., the optineurin E50K knock-in or the glutamate aspartate transporter (GLAST)-deficient mouse. Excitotoxicity can be mimicked by injecting the glutamate analogue N-methyl-D-aspartate intravitreally, which leads to rapid RGC degeneration. To explore the contribution of the immune system, the experimental autoimmune glaucoma model can serve as a useful tool. Here, immunization with antigens led to glaucoma-like damage. The ischemic mechanism can be mimicked by inducing a high IOP for a certain amount of time in rodents, followed by reperfusion. Thereby, damage to the retina and the optic nerve occurs rapidly after ischemia/reperfusion. Lastly, we discuss the importance of optic nerve crush models as model systems for normal-tension glaucoma. In summary, various glaucoma models beyond IOP increase can be utilized.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Stephanie C. Joachim
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, In der Schornau 23-25, 44892 Bochum, Germany; (T.T.); (S.R.); (L.D.); (N.K.); (H.B.D.)
| |
Collapse
|
3
|
Reinehr S, Girbig RM, Schulte KK, Theile J, Asaad MA, Fuchshofer R, Dick H, Joachim SC. Enhanced glaucomatous damage accompanied by glial response in a new multifactorial mouse model. Front Immunol 2023; 13:1017076. [PMID: 36733392 PMCID: PMC9887307 DOI: 10.3389/fimmu.2022.1017076] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 12/19/2022] [Indexed: 01/18/2023] Open
Abstract
Introduction Glaucoma is a complex, multifactorial neurodegenerative disease, which can lead to blindness if left untreated. It seems that, among others, immune processes, elevated intraocular pressure (IOP), or a combination of these factors are responsible for glaucomatous damage. Here, we combined two glaucoma models to examine if a combination of risk factors (IOP and immune response) results in a more severe damage of retinal ganglion cells (RGCs) and the optic nerves as well as an additional glia activation. Methods Six-week-old wildtype (WT+ONA) and βB1-Connective Tissue Growth Factor (CTGF) mice (CTGF+ONA) were immunized with 1 mg ONA (optic nerve antigen). A WT and a CTGF control group (CTGF) received sodium chloride instead. IOP was measured before and every two weeks after immunization. After six weeks, electroretinogram (ERG) measurements were performed. Then, retinae and optic nerves were processed for (immuno-) histology. Further, mRNA levels of corresponding genes in optic nerve and retina were analyzed via RT-qPCR. Results Six weeks after immunization, the IOP in CTGF and CTGF+ONA mice was increased. The optic nerve of CTGF+ONA animals displayed the most severe cell inflammation, demyelination, and macroglia activation. Fewer numbers of oligodendrocytes were only observed in WT+ONA optic nerves, while more apoptotic cells triggered by the extrinsic pathway could be revealed in all three glaucoma groups. The number of microglia/macrophages was not altered within the optic nerves of all groups. The loss of neuronal cells, especially RGCs was most pronounced in CTGF+ONA retinae in the central part and this was accompanied by an enhanced activation of microglia/macrophages. Also, Müller cell activation could be noted in CTGF and CTGF+ONA retinae. Discussion In this new model, an additive degeneration could be noted in optic nerves as well as in the number of RGCs. These results suggest a potential additive role of high IOP and immune factors in glaucoma development, which will aid for understanding this multifactorial disease more precisely in the future.
Collapse
Affiliation(s)
- Sabrina Reinehr
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany,*Correspondence: Sabrina Reinehr,
| | - Renée M. Girbig
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Kim K. Schulte
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Janine Theile
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | - M. Ali Asaad
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Rudolf Fuchshofer
- Institute of Human Anatomy and Embryology, University Regensburg, Regensburg, Germany
| | - H. Burkhard Dick
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Stephanie C. Joachim
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| |
Collapse
|
4
|
Molecular regulation of neuroinflammation in glaucoma: Current knowledge and the ongoing search for new treatment targets. Prog Retin Eye Res 2022; 87:100998. [PMID: 34348167 PMCID: PMC8803988 DOI: 10.1016/j.preteyeres.2021.100998] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 07/23/2021] [Accepted: 07/26/2021] [Indexed: 12/11/2022]
Abstract
Neuroinflammation relying on the inflammatory responses of glial cells has emerged as an impactful component of the multifactorial etiology of neurodegeneration in glaucoma. It has become increasingly evident that despite early adaptive and reparative features of glial responses, prolonged reactivity of the resident glia, along with the peripheral immune cells, create widespread toxicity to retinal ganglion cell (RGC) axons, somas, and synapses. As much as the synchronized responses of astrocytes and microglia to glaucoma-related stress or neuron injury, their bi-directional interactions are critical to build and amplify neuroinflammation and to dictate the neurodegenerative outcome. Although distinct molecular programs regulate somatic and axonal degeneration in glaucoma, inhibition of neurodegenerative inflammation can provide a broadly beneficial treatment strategy to rescue RGC integrity and function. Since inflammatory toxicity and mitochondrial dysfunction are converging etiological paths that can boost each other and feed into a vicious cycle, anti-inflammatory treatments may also offer a multi-target potential. This review presents an overview of the current knowledge on neuroinflammation in glaucoma with particular emphasis on the cell-intrinsic and cell-extrinsic factors involved in the reciprocal regulation of glial responses, the interdependence between inflammatory and mitochondrial routes of neurodegeneration, and the research aspects inspiring for prospective immunomodulatory treatments. With the advent of powerful technologies, ongoing research on molecular and functional characteristics of glial responses is expected to accumulate more comprehensive and complementary information and to rapidly move the field forward to safe and effective modulation of the glial pro-inflammatory activities, while restoring or augmenting the glial immune-regulatory and neurosupport functions.
Collapse
|
5
|
Hunziker D, Reinehr S, Palmhof M, Wagner N, Biniasch T, Stute G, Mattei P, Schmitz P, DiGiorgio P, Hert J, Rudolph MG, Benz J, Stihle M, Gsell B, Müller S, Gasser R, Schonhoven N, Ullmer C, Joachim SC. Synthesis, Characterization, and in vivo Evaluation of a Novel Potent Autotaxin-Inhibitor. Front Pharmacol 2022; 12:699535. [PMID: 35126098 PMCID: PMC8807399 DOI: 10.3389/fphar.2021.699535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 11/30/2021] [Indexed: 11/13/2022] Open
Abstract
The autotaxin-lysophosphatidic acid (ATX-LPA) signaling pathway plays a role in a variety of autoimmune diseases, such as rheumatoid arthritis or neurodegeneration. A link to the pathogenesis of glaucoma is suggested by an overactive ATX-LPA axis in aqueous humor samples of glaucoma patients. Analysis of such samples suggests that the ATX-LPA axis contributes to the fibrogenic activity and resistance to aqueous humor outflow through the trabecular meshwork. In order to inhibit or modulate this pathway, we developed a new series of ATX-inhibitors containing novel bicyclic and spirocyclic structural motifs. A potent lead compound (IC50 against ATX: 6 nM) with good in vivo PK, favorable in vitro property, and safety profile was generated. This compound leads to lowered LPA levels in vivo after oral administration. Hence, it was suitable for chronic oral treatment in two rodent models of glaucoma, the experimental autoimmune glaucoma (EAG) and the ischemia/reperfusion models. In the EAG model, rats were immunized with an optic nerve antigen homogenate, while controls received sodium chloride. Retinal ischemia/reperfusion (I/R) was induced by elevating the intraocular pressure (IOP) in one eye to 140 mmHg for 60 min, followed by reperfusion, while the other untreated eye served as control. Retinae and optic nerves were evaluated 28 days after EAG or 7 and 14 days after I/R induction. Oral treatment with the optimized ATX-inhibitor lead to reduced retinal ganglion cell (RGC) loss in both glaucoma models. In the optic nerve, the protective effect of ATX inhibition was less effective compared to the retina and only a trend to a weakened neurofilament distortion was detectable. Taken together, these results provide evidence that the dysregulation of the ATX-LPA axis in the aqueous humor of glaucoma patients, in addition to the postulated outflow impairment, might also contribute to RGC loss. The observation that ATX-inhibitor treatment in both glaucoma models did not result in significant IOP increases or decreases after oral treatment indicates that protection from RGC loss due to inhibition of the ATX-LPA axis is independent of an IOP lowering effect.
Collapse
Affiliation(s)
- Daniel Hunziker
- F. Hoffmann-La Roche Ltd., Pharma Research and Early Development, Therapeutic Modalities, Small Molecule Research, Roche Innovation Center Basel, Basel, Switzerland
| | - Sabrina Reinehr
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Marina Palmhof
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Natalie Wagner
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Thomas Biniasch
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Gesa Stute
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Patrizio Mattei
- F. Hoffmann-La Roche Ltd., Pharma Research and Early Development, Therapeutic Modalities, Small Molecule Research, Roche Innovation Center Basel, Basel, Switzerland
| | - Petra Schmitz
- F. Hoffmann-La Roche Ltd., Pharma Research and Early Development, Therapeutic Modalities, Small Molecule Research, Roche Innovation Center Basel, Basel, Switzerland
| | - Patrick DiGiorgio
- F. Hoffmann-La Roche Ltd., Pharma Research and Early Development, Therapeutic Modalities, Small Molecule Research, Roche Innovation Center Basel, Basel, Switzerland
| | - Jérôme Hert
- F. Hoffmann-La Roche Ltd., Pharma Research and Early Development, Therapeutic Modalities, Small Molecule Research, Roche Innovation Center Basel, Basel, Switzerland
| | - Markus G. Rudolph
- F. Hoffmann-La Roche Ltd., Pharma Research and Early Development, Therapeutic Modalities, Small Molecule Research, Roche Innovation Center Basel, Basel, Switzerland
| | - Joerg Benz
- F. Hoffmann-La Roche Ltd., Pharma Research and Early Development, Therapeutic Modalities, Small Molecule Research, Roche Innovation Center Basel, Basel, Switzerland
| | - Martine Stihle
- F. Hoffmann-La Roche Ltd., Pharma Research and Early Development, Therapeutic Modalities, Small Molecule Research, Roche Innovation Center Basel, Basel, Switzerland
| | - Bernard Gsell
- F. Hoffmann-La Roche Ltd., Pharma Research and Early Development, Therapeutic Modalities, Small Molecule Research, Roche Innovation Center Basel, Basel, Switzerland
| | - Stephan Müller
- F. Hoffmann-La Roche Ltd., Pharma Research and Early Development, Therapeutic Modalities, Small Molecule Research, Roche Innovation Center Basel, Basel, Switzerland
| | - Rodolfo Gasser
- F. Hoffmann-La Roche Ltd., Pharma Research and Early Development, Therapeutic Modalities, Small Molecule Research, Roche Innovation Center Basel, Basel, Switzerland
- F. Hoffmann-La Roche Ltd., Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, Basel, Switzerland
| | - Nina Schonhoven
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Christoph Ullmer
- F. Hoffmann-La Roche Ltd., Pharma Research and Early Development, Ophthalmology Discovery, Roche Innovation Center Basel, Basel, Switzerland
- *Correspondence: Christoph Ullmer, ; Stephanie C. Joachim,
| | - Stephanie C. Joachim
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
- *Correspondence: Christoph Ullmer, ; Stephanie C. Joachim,
| |
Collapse
|
6
|
Auler N, Tonner H, Pfeiffer N, Grus FH. Antibody and Protein Profiles in Glaucoma: Screening of Biomarkers and Identification of Signaling Pathways. BIOLOGY 2021; 10:biology10121296. [PMID: 34943212 PMCID: PMC8698915 DOI: 10.3390/biology10121296] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 11/24/2021] [Accepted: 12/03/2021] [Indexed: 12/12/2022]
Abstract
Simple Summary Glaucoma is a chronic eye disease that is one of the leading causes of blindness worldwide. Currently, the only therapeutic option is to lower intraocular pressure. The onset of the disease is often delayed because patients do not notice visual impairment until very late, which is why glaucoma is also known as “the silent thief of sight”. Therefore, early detection and definition of specific markers, the so-called biomarkers, are immensely important. For the methodical implementation, high-throughput methods and omic-based methods came more and more into focus. Thus, interesting targets for possible biomarkers were already suggested by clinical research and basic research, respectively. This review article aims to join the findings of the two disciplines by collecting overlaps as well as differences in various clinical studies and to shed light on promising candidates concerning findings from basic research, facilitating conclusions on possible therapy options. Abstract Glaucoma represents a group of chronic neurodegenerative diseases, constituting the second leading cause of blindness worldwide. To date, chronically elevated intraocular pressure has been identified as the main risk factor and the only treatable symptom. However, there is increasing evidence in the recent literature that IOP-independent molecular mechanisms also play an important role in the progression of the disease. In recent years, it has become increasingly clear that glaucoma has an autoimmune component. The main focus nowadays is elucidating glaucoma pathogenesis, finding early diagnostic options and new therapeutic approaches. This review article summarizes the impact of different antibodies and proteins associated with glaucoma that can be detected for example by microarray and mass spectrometric analyzes, which (i) provide information about expression profiles and associated molecular signaling pathways, (ii) can possibly be used as a diagnostic tool in future and, (iii) can identify possible targets for therapeutic approaches.
Collapse
|
7
|
Sharif NA. Therapeutic Drugs and Devices for Tackling Ocular Hypertension and Glaucoma, and Need for Neuroprotection and Cytoprotective Therapies. Front Pharmacol 2021; 12:729249. [PMID: 34603044 PMCID: PMC8484316 DOI: 10.3389/fphar.2021.729249] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 08/18/2021] [Indexed: 12/11/2022] Open
Abstract
Damage to the optic nerve and the death of associated retinal ganglion cells (RGCs) by elevated intraocular pressure (IOP), also known as glaucoma, is responsible for visual impairment and blindness in millions of people worldwide. The ocular hypertension (OHT) and the deleterious mechanical forces it exerts at the back of the eye, at the level of the optic nerve head/optic disc and lamina cribosa, is the only modifiable risk factor associated with glaucoma that can be treated. The elevated IOP occurs due to the inability of accumulated aqueous humor (AQH) to egress from the anterior chamber of the eye due to occlusion of the major outflow pathway, the trabecular meshwork (TM) and Schlemm’s canal (SC). Several different classes of pharmaceutical agents, surgical techniques and implantable devices have been developed to lower and control IOP. First-line drugs to promote AQH outflow via the uveoscleral outflow pathway include FP-receptor prostaglandin (PG) agonists (e.g., latanoprost, travoprost and tafluprost) and a novel non-PG EP2-receptor agonist (omidenepag isopropyl, Eybelis®). TM/SC outflow enhancing drugs are also effective ocular hypotensive agents (e.g., rho kinase inhibitors like ripasudil and netarsudil; and latanoprostene bunod, a conjugate of a nitric oxide donor and latanoprost). One of the most effective anterior chamber AQH microshunt devices is the Preserflo® microshunt which can lower IOP down to 10–13 mmHg. Other IOP-lowering drugs and devices on the horizon will be also discussed. Additionally, since elevated IOP is only one of many risk factors for development of glaucomatous optic neuropathy, a treatise of the role of inflammatory neurodegeneration of the optic nerve and retinal ganglion cells and appropriate neuroprotective strategies to mitigate this disease will also be reviewed and discussed.
Collapse
Affiliation(s)
- Najam A Sharif
- Global Alliances and External Research, Ophthalmology Innovation Center, Santen Inc., Emeryville, CA, United States
| |
Collapse
|
8
|
Yu L, Chen Y, Xu X, Dong Q, Xiu W, Chen Q, Wang J, He C, Ye J, Lu F. Alterations in Peripheral B Cell Subsets Correlate with the Disease Severity of Human Glaucoma. J Inflamm Res 2021; 14:4827-4838. [PMID: 34584441 PMCID: PMC8464325 DOI: 10.2147/jir.s329084] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 09/16/2021] [Indexed: 12/13/2022] Open
Abstract
Background Glaucoma is a group of retinal neurodegenerative diseases causing irreversible visual impairment. The pathogenesis of this disease is complicated. Studies have shown that the immune system is involved in the neurodegenerative process of glaucoma. There are continuous evidences that autoantibodies play a crucial role in the pathogenesis of glaucoma. However, focuses on B cells, the antibody-producing cells in glaucoma are surprisingly limited. Methods Fresh peripheral blood samples were collected from 44 glaucoma patients (38 with primary angle-closure glaucoma (PACG) and 6 with (primary open-angle glaucoma POAG)) and 36 age-matched healthy donors (HD). Density gradient centrifugation was performed to obtain peripheral blood mononuclear cells (PBMC). Flow cytometry was performed to determine B cell phenotypes. The severity of glaucoma was determined based on the mean deviation (MD) of visual field. Results In this study, we demonstrated that total B cells was significantly increased in glaucoma patients compared to HD. Next, we checked changes of different B cell subsets in glaucoma. Glaucoma patients were found to have a significant increase in the frequencies of antibody-secreting cells (ASC)/plasmablasts, naïve, and CD19+ CD27− IgD− double negative (DN) subpopulations, but a decrease in the CD27+ IgD+ unswitched memory compartment. Notably, we found that the increment of CD27− IgD− DN B cells was significantly magnified according to the clinical severity. Conclusion We demonstrate, for the first time, that peripheral B cell subsets are altered and unveil the correlation of a newly identified pro-inflammatory CD27− IgD− DN subset with clinical features of glaucoma, suggesting that these B cell subsets could serve as potential biomarkers to monitor the disease progression of glaucoma patients.
Collapse
Affiliation(s)
- Ling Yu
- Department of Ophthalmology, Daping Hospital, Army Medical Center, Army Medical University, Chongqing, People's Republic of China
| | - Yang Chen
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, People's Republic of China
| | - Xiang Xu
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, People's Republic of China
| | - Qiwei Dong
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, People's Republic of China.,Medico-Engineering Cooperation on Applied Medicine Research Center, University of Electronic Science and Technology of China, Chengdu, People's Republic of China
| | - Wenbo Xiu
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, People's Republic of China
| | - Qinyuan Chen
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, People's Republic of China
| | - Jinxia Wang
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, People's Republic of China
| | - Chong He
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, People's Republic of China.,Medico-Engineering Cooperation on Applied Medicine Research Center, University of Electronic Science and Technology of China, Chengdu, People's Republic of China
| | - Jian Ye
- Department of Ophthalmology, Daping Hospital, Army Medical Center, Army Medical University, Chongqing, People's Republic of China
| | - Fang Lu
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, People's Republic of China.,Medico-Engineering Cooperation on Applied Medicine Research Center, University of Electronic Science and Technology of China, Chengdu, People's Republic of China
| |
Collapse
|
9
|
Duarte JN. Neuroinflammatory Mechanisms of Mitochondrial Dysfunction and Neurodegeneration in Glaucoma. J Ophthalmol 2021; 2021:4581909. [PMID: 33953963 PMCID: PMC8064803 DOI: 10.1155/2021/4581909] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 06/29/2020] [Accepted: 03/23/2021] [Indexed: 12/13/2022] Open
Abstract
The exact mechanism of retinal ganglion cell loss in the pathogenesis of glaucoma is yet to be understood. Mitochondrial damage-associated molecular patterns (DAMPs) resulting from mitochondrial dysfunction have been linked to Leber's hereditary optic neuropathy and autosomal dominant optic atrophy, as well as to brain neurodegenerative diseases. Recent evidence shows that, in conditions where mitochondria are damaged, a sustained inflammatory response and downstream pathological inflammation may ensue. Mitochondrial damage has been linked to the accumulation of age-related mitochondrial DNA mutations and mitochondrial dysfunction, possibly through aberrant reactive oxygen species production and defective mitophagy. The present review focuses on how mitochondrial dysfunction may overwhelm the ability of neurons and glial cells to adequately maintain homeostasis and how mitochondria-derived DAMPs trigger the immune system and induce neurodegeneration.
Collapse
Affiliation(s)
- Joao N. Duarte
- Neuroinflammation Unit, Biotech Research & Innovation Center, University of Copenhagen, Copenhagen, Denmark
- Department of Ophthalmology, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Immunology, Section 7631, Rigshospitalet, Copenhagen, Denmark
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
10
|
Tribble JR, Kokkali E, Otmani A, Plastino F, Lardner E, Vohra R, Kolko M, André H, Morgan JE, Williams PA. When Is a Control Not a Control? Reactive Microglia Occur Throughout the Control Contralateral Pathway of Retinal Ganglion Cell Projections in Experimental Glaucoma. Transl Vis Sci Technol 2021; 10:22. [PMID: 33510961 PMCID: PMC7804521 DOI: 10.1167/tvst.10.1.22] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 12/02/2020] [Indexed: 12/19/2022] Open
Abstract
Purpose Animal models show retinal ganglion cell (RGC) injuries that replicate features of glaucoma and the contralateral eye is commonly used as an internal control. There is significant crossover of RGC axons from the ipsilateral to the contralateral side at the level of the optic chiasm, which may confound findings when damage is restricted to one eye. The effect of unilateral glaucoma on neuroinflammatory damage to the contralateral pathway of RGC projections has largely been unexplored. Methods Ocular hypertensive glaucoma was induced unilaterally or bilaterally in the rat and RGC neurodegenerative events were assessed. Neuroinflammation was quantified in the retina, optic nerve head, optic nerve, lateral geniculate nucleus, and superior colliculus by high-resolution imaging, and in the retina by flow cytometry and protein arrays. Results After ocular hypertensive stress, peripheral monocytes enter the retina and microglia become reactive. This effect is more marked in animals with bilateral ocular hypertensive glaucoma. In rats where glaucoma was induced unilaterally, there was significant microglia activation in the contralateral (control) eye. Microglial activation extended into the optic nerve and terminal visual thalami, where it was similar across hemispheres in unilateral ocular hypertension. Conclusions These data suggest that caution is warranted when using the contralateral eye as a control and in comparing visual thalami in unilateral models of glaucoma. Translational Relevance The use of a contralateral eye as a control may confound the discovery of human-relevant mechanism and treatments in animal models. We also identify neuroinflammatory protein responses that warrant further investigation as potential disease-modifiable targets.
Collapse
Affiliation(s)
- James R. Tribble
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Eirini Kokkali
- School of Optometry and Vision Sciences, Cardiff University, Cardiff, Wales, UK
| | - Amin Otmani
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Flavia Plastino
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Emma Lardner
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Rupali Vohra
- Department of Veterinary and Animal Sciences, Pathobiological Sciences, University of Copenhagen, Denmark
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Miriam Kolko
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
- Department of Ophthalmology, Rigshospitalet-Glostrup, Copenhagen, Denmark
| | - Helder André
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - James E. Morgan
- School of Optometry and Vision Sciences, Cardiff University, Cardiff, Wales, UK
- School of Medicine, Cardiff University, Cardiff, Wales, UK
| | - Pete A. Williams
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
11
|
Hurst J, Fietz A, Tsai T, Joachim SC, Schnichels S. Organ Cultures for Retinal Diseases. Front Neurosci 2020; 14:583392. [PMID: 33324149 PMCID: PMC7724035 DOI: 10.3389/fnins.2020.583392] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 10/13/2020] [Indexed: 12/18/2022] Open
Abstract
The successful development of novel therapies is closely linked with understanding the underlying pathomechanisms of a disease. To do so, model systems that reflect human diseases and allow for the evaluation of new therapeutic approaches are needed. Yet, preclinical animal studies often have limited success in predicting human physiology, pathology, and therapeutic responses. Moreover, animal testing is facing increasing ethical and bureaucratic hurdles, while human cell cultures are limited in their ability to represent in vivo situations due to the lack of the tissue microenvironment, which may alter cellular responses. To overcome these struggles, organ cultures, especially those of complex organs such as the retina, can be used to study physiological reactions to substances or stressors. Human and animal organ cultures are now well established and recognized. This mini-review discusses how retinal organ cultures can be used to preserve tissue architecture more realistically and therefore better represent disease-related changes. It also shows how molecular biological, biochemical, and histological techniques can be combined to investigate how anatomical localization may alter cellular responses. Examples for the use of retinal organ cultures, including models to study age-related macular degeneration (AMD), retinitis pigmentosa (RP), central artery occlusion (CRAO), and glaucoma are presented, and their advantages and disadvantages are discussed. We conclude that organ cultures significantly improve our understanding of complex retinal diseases and may advance treatment testing without the need for animal testing.
Collapse
Affiliation(s)
- José Hurst
- Center for Ophthalmology, University Eye Hospital, University of Tübingen, Tübingen, Germany
| | - Agnes Fietz
- Center for Ophthalmology, University Eye Hospital, University of Tübingen, Tübingen, Germany
| | - Teresa Tsai
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Stephanie C. Joachim
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Sven Schnichels
- Center for Ophthalmology, University Eye Hospital, University of Tübingen, Tübingen, Germany
| |
Collapse
|
12
|
Reinehr S, Buschhorn V, Mueller-Buehl AM, Goldmann T, Grus FH, Wolfrum U, Dick HB, Joachim SC. Occurrence of Retinal Ganglion Cell Loss via Autophagy and Apoptotic Pathways in an Autoimmune Glaucoma Model. Curr Eye Res 2020; 45:1124-1135. [PMID: 31935132 DOI: 10.1080/02713683.2020.1716987] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 01/01/2020] [Accepted: 01/07/2020] [Indexed: 12/17/2022]
Abstract
PURPOSE In glaucoma, an apoptotic death of retinal ganglion cells (RGCs) has been shown. However, little is known about other cell death mechanisms, like autophagy or necrosis. Therefore, we investigated these mechanisms in addition to antibody deposits in an experimental autoimmune glaucoma model. METHODS Rats were immunized with a retinal ganglion cell-layer homogenate (RGA), while controls received sodium chloride. Untreated rats served as natїve group. After seven weeks, retinal cross-sections were stained with antibodies against RGCs (Brn-3a), apoptosis (cleaved caspase 2, cleaved caspase 3 as well as caspase 3, 8, and 9), autophagy (LC3BII and LAMP1), and necrosis (RIPK3) followed by cell counts. Autophagy was additionally visualized via transmission electron microscopy on retinal sections. Antibody deposits were also analyzed. RESULTS We noted a RGC loss after RGA immunization compared to both control groups. Also, significantly more cleaved caspase 2+ RGCs were observed in RGA animals. More caspase 3 and 8 signals were noted in RGA retinas compared to both controls, while no changes were seen in regard to caspase 9. Furthermore, significantly more cleaved caspase 3+ cells were detected in RGA animals. We noted an increase of LC3BII+ and LAMP1+ autophagic cells in the RGA group, while no alterations were seen regarding necrotic RIPK3+ cells. Autophagic vesicles were observed via transmission electron microscopy. IgG staining revealed significant differences between the RGA group and controls concerning IgG deposits in the ganglion cell layer. CONCLUSIONS Due to the novel results from this study, we conclude that IgG antibodies are involved in RGC loss in this model leading to apoptotic and autophagic cell loss. These results could help to develop new therapy strategies for glaucoma patients.
Collapse
Affiliation(s)
- Sabrina Reinehr
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum , Bochum, Germany
| | - Verena Buschhorn
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum , Bochum, Germany
| | - Ana M Mueller-Buehl
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum , Bochum, Germany
| | - Tobias Goldmann
- Molecular Cell Biology, Institute of Molecular Physiology, Johannes Gutenberg-University of Mainz , Germany
| | - Franz H Grus
- Experimental Ophthalmology, University Medical Center Mainz , Mainz, Germany
| | - Uwe Wolfrum
- Molecular Cell Biology, Institute of Molecular Physiology, Johannes Gutenberg-University of Mainz , Germany
| | - H Burkhard Dick
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum , Bochum, Germany
| | - Stephanie C Joachim
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum , Bochum, Germany
| |
Collapse
|
13
|
Edwards G, Perkins GA, Kim KY, Kong Y, Lee Y, Choi SH, Liu Y, Skowronska-Krawczyk D, Weinreb RN, Zangwill L, Strack S, Ju WK. Loss of AKAP1 triggers Drp1 dephosphorylation-mediated mitochondrial fission and loss in retinal ganglion cells. Cell Death Dis 2020; 11:254. [PMID: 32312949 PMCID: PMC7170863 DOI: 10.1038/s41419-020-2456-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 04/06/2020] [Accepted: 04/07/2020] [Indexed: 12/14/2022]
Abstract
Impairment of mitochondrial structure and function is strongly linked to glaucoma pathogenesis. Despite the widely appreciated disease relevance of mitochondrial dysfunction and loss, the molecular mechanisms underlying mitochondrial fragmentation and metabolic stress in glaucoma are poorly understood. We demonstrate here that glaucomatous retinal ganglion cells (RGCs) show loss of A-kinase anchoring protein 1 (AKAP1), activation of calcineurin (CaN) and reduction of dynamin-related protein 1 (Drp1) phosphorylation at serine 637 (Ser637). These findings suggest that AKAP1-mediated phosphorylation of Drp1 at Ser637 has a critical role in RGC survival in glaucomatous neurodegeneration. Male mice lacking AKAP1 show increases in CaN and total Drp1 levels, as well as a decrease in Drp1 phosphorylation at Ser637 in the retina. Ultrastructural analysis of mitochondria shows that loss of AKAP1 triggers mitochondrial fragmentation and loss, as well as mitophagosome formation in RGCs. Loss of AKAP1 deregulates oxidative phosphorylation (OXPHOS) complexes (Cxs) by increasing CxII and decreasing CxIII-V, leading to metabolic and oxidative stress. Also, loss of AKAP1 decreases Akt phosphorylation at Serine 473 (Ser473) and threonine 308 (Thr308) and activates the Bim/Bax signaling pathway in the retina. These results suggest that loss of AKAP1 has a critical role in RGC dysfunction by decreasing Drp1 phosphorylation at Ser637, deregulating OXPHOS, decreasing Akt phosphorylation at Ser473 and Thr308, and activating the Bim/Bax pathway in glaucomatous neurodegeneration. Thus, we propose that overexpression of AKAP1 or modulation of Drp1 phosphorylation at Ser637 are potential therapeutic strategies for neuroprotective intervention in glaucoma and other mitochondria-related optic neuropathies.
Collapse
Affiliation(s)
- Genea Edwards
- Hamilton Glaucoma Center and Shiley Eye Center, The Viterbi Family Department of Ophthalmology, University of California San Diego, La Jolla, CA, USA
| | - Guy A Perkins
- National Center for Microscopy and Imaging Research and Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Keun-Young Kim
- National Center for Microscopy and Imaging Research and Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - YeEun Kong
- Hamilton Glaucoma Center and Shiley Eye Center, The Viterbi Family Department of Ophthalmology, University of California San Diego, La Jolla, CA, USA
| | - Yonghoon Lee
- Hamilton Glaucoma Center and Shiley Eye Center, The Viterbi Family Department of Ophthalmology, University of California San Diego, La Jolla, CA, USA
| | - Soo-Ho Choi
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Yujia Liu
- Department of Pharmacology and Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
| | - Dorota Skowronska-Krawczyk
- Hamilton Glaucoma Center and Shiley Eye Center, The Viterbi Family Department of Ophthalmology, University of California San Diego, La Jolla, CA, USA
| | - Robert N Weinreb
- Hamilton Glaucoma Center and Shiley Eye Center, The Viterbi Family Department of Ophthalmology, University of California San Diego, La Jolla, CA, USA
| | - Linda Zangwill
- Hamilton Glaucoma Center and Shiley Eye Center, The Viterbi Family Department of Ophthalmology, University of California San Diego, La Jolla, CA, USA
| | - Stefan Strack
- Department of Pharmacology and Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
| | - Won-Kyu Ju
- Hamilton Glaucoma Center and Shiley Eye Center, The Viterbi Family Department of Ophthalmology, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
14
|
Nor Arfuzir NN, Agarwal R, Iezhitsa I, Agarwal P, Ismail NM. Magnesium acetyltaurate protects against endothelin-1 induced RGC loss by reducing neuroinflammation in Sprague dawley rats. Exp Eye Res 2020; 194:107996. [PMID: 32156652 DOI: 10.1016/j.exer.2020.107996] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 02/25/2020] [Accepted: 03/06/2020] [Indexed: 12/22/2022]
Abstract
Endothelin-1 (ET-1), a potent vasoconstrictor, plays a significant role in the pathophysiology of ocular conditions like glaucoma. Glaucoma is characterized by apoptotic loss of retinal ganglion cells (RGCs) and loss of visual fields and is a leading cause of irreversible blindness. In glaucomatous eyes, retinal ischemia causes release of pro-inflammatory mediators such as interleukin (IL)-1β, IL-6 and tumor necrosis factor (TNF)-α and promotes activation of transcription factors such as nuclear factor kappa B (NFKB) and c-Jun. Magnesium acetyltaurate (MgAT) has previously been shown to protect against ET-1 induced retinal and optic nerve damage. Current study investigated the mechanisms underlying these effects of MgAT, which so far remain unknown. Sprague dawley rats were intravitreally injected with ET-1 with or without pretreatment with MgAT. Seven days post-injection, retinal expression of IL-1β, IL-6, TNF-α, NFKB and c-Jun protein and genes was determined using multiplex assay, Western blot and PCR. Animals were subjected to retrograde labeling of RGCs to determine the extent of RGC survival. RGC survival was also examined using Brn3A staining. Furthermore, visual functions of rats were determined using Morris water maze. It was observed that pre-treatment with MgAT protects against ET-1 induced increase in the retinal expression of IL-1β, IL-6 and TNF-α proteins and genes. It also protected against ET-1 induced activation of NFKB and c-Jun. These effects of MgAT were associated with greater RGC survival and preservation of visual functions in rats. In conclusion, MgAT prevents ET-1 induced RGC loss and loss of visual functions by suppressing neuroinflammatory reaction in rat retinas.
Collapse
Affiliation(s)
- Natasha Najwa Nor Arfuzir
- Center for Neuroscience Research, Faculty of Medicine, Universiti Teknologi MARA Sungai Buloh Campus, Selangor, Malaysia
| | - Renu Agarwal
- School of Medicine, International Medical University, Kuala Lumpur, Malaysia.
| | - Igor Iezhitsa
- Center for Neuroscience Research, Faculty of Medicine, Universiti Teknologi MARA Sungai Buloh Campus, Selangor, Malaysia; Volgograd State Medical University, Research Centre for Innovative Medicines, Volgograd, Russian Federation; Institute for Pathology, Laboratory and Forensic Medicine (I-PPerForM), Universiti Teknologi MARA, Sungai Buloh Campus, Selangor, Malaysia
| | - Puneet Agarwal
- School of Medicine, International Medical University, Kuala Lumpur, Malaysia
| | - Nafeeza Mohd Ismail
- School of Medicine, International Medical University, Kuala Lumpur, Malaysia
| |
Collapse
|
15
|
Grotegut P, Kuehn S, Meißner W, Dick HB, Joachim SC. Intravitreal S100B Injection Triggers a Time-Dependent Microglia Response in a Pro-Inflammatory Manner in Retina and Optic Nerve. Mol Neurobiol 2019; 57:1186-1202. [DOI: 10.1007/s12035-019-01786-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 09/12/2019] [Indexed: 12/14/2022]
|
16
|
Tsai T, Reinehr S, Maliha AM, Joachim SC. Immune Mediated Degeneration and Possible Protection in Glaucoma. Front Neurosci 2019; 13:931. [PMID: 31543759 PMCID: PMC6733056 DOI: 10.3389/fnins.2019.00931] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 08/19/2019] [Indexed: 12/13/2022] Open
Abstract
The underlying pathomechanisms for glaucoma, one of the most common causes of blindness worldwide, are still not identified. In addition to increased intraocular pressure (IOP), oxidative stress, excitotoxicity, and immunological processes seem to play a role. Several pharmacological or molecular/genetic methods are currently investigated as treatment options for this disease. Altered autoantibody levels were detected in serum, aqueous humor, and tissue sections of glaucoma patients. To further analyze the role of the immune system, an IOP-independent, experimental autoimmune glaucoma (EAG) animal model was developed. In this model, immunization with ocular antigens leads to antibody depositions, misdirected T-cells, retinal ganglion cell death and degeneration of the optic nerve, similar to glaucomatous degeneration in patients. Moreover, an activation of the complement system and microglia alterations were identified in the EAG as well as in ocular hypertension models. The inhibition of these factors can alleviate degeneration in glaucoma models with and without high IOP. Currently, several neuroprotective approaches are tested in distinct models. It is necessary to have systems that cover underlying pathomechanisms, but also allow for the screening of new drugs. In vitro models are commonly used, including single cell lines, mixed-cultures, and even organoids. In ex vivo organ cultures, pathomechanisms as well as therapeutics can be investigated in the whole retina. Furthermore, animal models reveal insights in the in vivo situation. With all these models, several possible new drugs and therapy strategies were tested in the last years. For example, hypothermia treatment, neurotrophic factors or the blockage of excitotoxity. However, further studies are required to reveal the pressure independent pathomechanisms behind glaucoma. There is still an open issue whether immune mechanisms directly or indirectly trigger cell death pathways. Hence, it might be an imbalance between protective and destructive immune mechanisms. Moreover, identified therapy options have to be evaluated in more detail, since deeper insights could lead to better treatment options for glaucoma patients.
Collapse
Affiliation(s)
| | | | | | - Stephanie C. Joachim
- Experimental Eye Research, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| |
Collapse
|
17
|
Yang X, Zeng Q, Göktas E, Gopal K, Al-Aswad L, Blumberg DM, Cioffi GA, Liebmann JM, Tezel G. T-Lymphocyte Subset Distribution and Activity in Patients With Glaucoma. Invest Ophthalmol Vis Sci 2019; 60:877-888. [PMID: 30821813 PMCID: PMC6397017 DOI: 10.1167/iovs.18-26129] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Purpose Besides glia-driven neuroinflammation, growing evidence from analysis of human blood samples, isolated autoantibodies, and postmortem tissues also support systemic immune responses during neurodegeneration in glaucoma patients. To explore the T-cell–mediated component of systemic immunity, this study analyzed T lymphocytes in patients' blood. Methods Blood samples were collected from 32 patients with glaucoma and 21 nonglaucomatous controls, and mononuclear cells were isolated by Histopaque density gradient centrifugation. T-cell subset distribution was analyzed by multicolor flow cytometry after helper (Th) and cytotoxic fractions, and Th subpopulations, were stained with antibodies to CD4, CD8, or distinctive markers, such as IFN-γ (for Th1), IL-4 (for Th2), IL-17A (for Th17), and CD25/FoxP3 (for T regulatory cells [Tregs]). In addition, proliferative activity and cytokine secretion of T cells were analyzed after in vitro stimulation. Results Analysis of T-cell subset distribution detected a glaucoma-related shift. Despite similar frequencies of CD4+ or CD8+ T cells, or Th1, Th2, or Th17 subsets in glaucoma and control groups, glaucomatous samples exhibited a trend toward decreased frequency of CD4+ (or CD8+)/CD25+/FoxP3+ Tregs within the entire CD4+ (or CD8+) population (P < 0.001). Furthermore, CD4+ T cells in glaucomatous samples presented a greater stimulation response (∼3-fold) as characterized by increased proliferation and proinflammatory cytokine secretion (P < 0.05). Conclusions These findings suggest that the immunity activated in glaucoma may not be counterbalanced by an efficient immune suppression. More work is encouraged to determine whether shifted T-cell homeostasis may contribute to neurodegeneration in glaucoma, and/or whether T-cell subset imbalance may serve as a biomarker of autoimmune susceptibility.
Collapse
Affiliation(s)
- Xiangjun Yang
- Department of Ophthalmology, Columbia University, Vagelos College of Physicians and Surgeons, New York, New York, United States
| | - Qun Zeng
- Department of Ophthalmology, Columbia University, Vagelos College of Physicians and Surgeons, New York, New York, United States
| | - Emre Göktas
- Department of Ophthalmology, Columbia University, Vagelos College of Physicians and Surgeons, New York, New York, United States
| | - Kalashree Gopal
- Department of Ophthalmology, Columbia University, Vagelos College of Physicians and Surgeons, New York, New York, United States
| | - Lama Al-Aswad
- Department of Ophthalmology, Columbia University, Vagelos College of Physicians and Surgeons, New York, New York, United States
| | - Dana M Blumberg
- Department of Ophthalmology, Columbia University, Vagelos College of Physicians and Surgeons, New York, New York, United States
| | - George A Cioffi
- Department of Ophthalmology, Columbia University, Vagelos College of Physicians and Surgeons, New York, New York, United States
| | - Jeffrey M Liebmann
- Department of Ophthalmology, Columbia University, Vagelos College of Physicians and Surgeons, New York, New York, United States
| | - Gülgün Tezel
- Department of Ophthalmology, Columbia University, Vagelos College of Physicians and Surgeons, New York, New York, United States
| |
Collapse
|
18
|
Reinehr S, Koch D, Weiss M, Froemel F, Voss C, Dick HB, Fuchshofer R, Joachim SC. Loss of retinal ganglion cells in a new genetic mouse model for primary open-angle glaucoma. J Cell Mol Med 2019; 23:5497-5507. [PMID: 31144440 PMCID: PMC6653331 DOI: 10.1111/jcmm.14433] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 03/18/2019] [Accepted: 05/15/2019] [Indexed: 12/14/2022] Open
Abstract
Primary open-angle glaucoma (POAG) is one of the most common causes for blindness worldwide. Although an elevated intraocular pressure (IOP) is the main risk factor, the exact pathology remained indistinguishable. Therefore, it is necessary to have appropriate models to investigate these mechanisms. Here, we analysed a transgenic glaucoma mouse model (βB1-CTGF) to elucidate new possible mechanisms of the disease. Therefore, IOP was measured in βB1-CTGF and wildtype mice at 5, 10 and 15 weeks of age. At 5 and 10 weeks, the IOP in both groups were comparable (P > 0.05). After 15 weeks, a significant elevated IOP was measured in βB1-CTGF mice (P < 0.001). At 15 weeks, electroretinogram measurements were performed and both the a- and b-wave amplitudes were significantly decreased in βB1-CTGF retinae (both P < 0.01). Significantly fewer Brn-3a+ retinal ganglion cells (RGCs) were observed in the βB1-CTGF group on flatmounts (P = 0.02), cross-sections (P < 0.001) and also via quantitative real-time PCR (P = 0.02). Additionally, significantly more cleaved caspase 3+ RGCs were seen in the βB1-CTGF group (P = 0.002). Furthermore, a decrease in recoverin+ cells was observable in the βB1-CTGF animals (P = 0.004). Accordingly, a significant down-regulation of Recoverin mRNA levels were noted (P < 0.001). Gfap expression, on the other hand, was higher in βB1-CTGF retinae (P = 0.023). Additionally, more glutamine synthetase signal was noted (P = 0.04). Although no alterations were observed regarding photoreceptors via immunohistology, a significant decrease of Rhodopsin (P = 0.003) and Opsin mRNA (P = 0.03) was noted. We therefore assume that the βB1-CTGF mouse could serve as an excellent model for better understanding the pathomechanisms in POAG.
Collapse
Affiliation(s)
- Sabrina Reinehr
- Experimental Eye Research InstituteUniversity Eye Hospital, Ruhr‐University BochumBochumGermany
| | - Dennis Koch
- Experimental Eye Research InstituteUniversity Eye Hospital, Ruhr‐University BochumBochumGermany
| | - Maximilian Weiss
- Experimental Eye Research InstituteUniversity Eye Hospital, Ruhr‐University BochumBochumGermany
| | - Franziska Froemel
- Institute of Human Anatomy and EmbryologyUniversity RegensburgRegensburgGermany
| | - Christina Voss
- Experimental Eye Research InstituteUniversity Eye Hospital, Ruhr‐University BochumBochumGermany
| | - H. Burkhard Dick
- Experimental Eye Research InstituteUniversity Eye Hospital, Ruhr‐University BochumBochumGermany
| | - Rudolf Fuchshofer
- Institute of Human Anatomy and EmbryologyUniversity RegensburgRegensburgGermany
| | - Stephanie C. Joachim
- Experimental Eye Research InstituteUniversity Eye Hospital, Ruhr‐University BochumBochumGermany
| |
Collapse
|
19
|
Abstract
In addition to the clinically most relevant risk factor for glaucoma, i.e., elevated intraocular pressure (IOP), there are other factors with high relevance for the disease. Changes in the autoimmune component of the immune system are of particular importance. Clinical studies have demonstrated alterations in different autoantibodies in glaucoma patients compared to healthy controls, some of which increase in abundance/have a raised titer, but also some which have a reduced titer. These changes have a distinct potential-not only as a tool for early glaucoma detection, but also as a therapeutic option due to the documented neuroprotective effects of some of these antibodies. Several antibodies displaying lower abundance in glaucoma patients, e.g., antibodies against 14-3-3 proteins, γ‑/α-synuclein, or also against glial fibrillary acidic protein (GFAP), show neuroprotective effects on retinal ganglion cells in vivo and in vitro. To assess the relevance of changes detected in the immune system of glaucoma patients, "‑omics-based" analyses of different ocular tissues are of particular importance alongside cell culture studies. In this manner, not only samples derived from experimental studies but also samples derived from glaucoma patients in even very small amounts (e. g., tears, aqueous humor, serum, or post-mortem retina) can be analyzed in detail in terms of protein and, in particular, antibody changes. Modern mass spectrometric proteomic characterization of relevant samples will deliver valuable information concerning the understanding of molecular disease mechanisms in the coming years, thus also improving diagnosis and treatment of glaucoma.
Collapse
Affiliation(s)
- K Bell
- Experimentelle Ophthalmologie, Augenklinik der Universitätsmedizin Mainz, Langenbeckstraße 1, 55131, Mainz, Deutschland.
| | - S Funke
- Experimentelle Ophthalmologie, Augenklinik der Universitätsmedizin Mainz, Langenbeckstraße 1, 55131, Mainz, Deutschland
| | - F H Grus
- Experimentelle Ophthalmologie, Augenklinik der Universitätsmedizin Mainz, Langenbeckstraße 1, 55131, Mainz, Deutschland
| |
Collapse
|
20
|
Bariş M, Tezel G. Immunomodulation as a Neuroprotective Strategy for Glaucoma Treatment. CURRENT OPHTHALMOLOGY REPORTS 2019; 7:160-169. [PMID: 31360618 PMCID: PMC6662642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
PURPOSE OF REVIEW This review aims to highlight the current knowledge about inflammatory mechanisms of neurodegeneration in glaucoma with emphasis on potential immunomodulation strategies. RECENT FINDINGS Glaucomatous retina and optic nerve present multiple evidences of inflammatory responses of astroglia, microglia, and blood-born immune cells. Although adaptive/protective responses of resident or systemic immune cells can support neurons and promote tissue repair mechanisms after injurious insults, prolonged inflammatory processes can also produce neurotoxic mediators. Treatments targeting these neurodestructive outcomes may restore immune homeostasis and protect neurons from inflammatory injury. Due to widespread and chronic nature of neuroinflammation in glaucoma, immunomodulation offers a treatment strategy to protect different neuronal compartments of RGCs during the chronic and asynchronous course of neurodegeneration. Uncovering of distinct molecular responses and interactions of different immune cells that determine the neuroinflammatory phenotype and participate in neurodegenerative outcomes will be critical to develop effective strategies for immunomodulation in glaucoma. SUMMARY Neuroinflammation has increasingly been recognized to play an important role in glaucomatous neurodegeneration, and its modulation appears to be a promising treatment strategy for neuroprotection.
Collapse
Affiliation(s)
- Mine Bariş
- Columbia University, College of Physicians and Surgeons, Department of Ophthalmology, New York, NY
| | - Gülgün Tezel
- Columbia University, College of Physicians and Surgeons, Department of Ophthalmology, New York, NY
| |
Collapse
|
21
|
Transfer of the Experimental Autoimmune Glaucoma Model from Rats to Mice-New Options to Study Glaucoma Disease. Int J Mol Sci 2019; 20:ijms20102563. [PMID: 31137749 PMCID: PMC6566658 DOI: 10.3390/ijms20102563] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 05/13/2019] [Accepted: 05/21/2019] [Indexed: 12/25/2022] Open
Abstract
Studies have suggested an involvement of the immune system in glaucoma. Hence, a rat experimental autoimmune glaucoma model (EAG) was developed to investigate the role of the immune response. Here, we transferred this model into mice. Either 0.8 mg/mL of the optic nerve antigen homogenate (ONA; ONA 0.8) or 1.0 mg/mL ONA (ONA 1.0) were injected in 129/Sv mice. Controls received sodium chloride. Before and 6 weeks after immunization, the intraocular pressure (IOP) was measured. At 6 weeks, retinal neurons, glia cells, and synapses were analyzed via immunohistology and quantitative real-time PCR (RT-qPCR). Additionally, optic nerves were examined. The IOP stayed in the normal physiological range throughout the study (p > 0.05). A significant reduction of retinal ganglion cells (RGCs) was noted in both immunized groups (p < 0.001). Remodeling of glutamatergic and GABAergic synapses was seen in ONA 1.0 retinas. Furthermore, both ONA groups revealed optic nerve degeneration and macrogliosis (all: p < 0.001). An increase of activated microglia was noted in ONA retinas and optic nerves (p < 0.05). Both ONA concentrations led to RGC loss and optic nerve degeneration. Therefore, the EAG model was successfully transferred from rats to mice. In further studies, transgenic knockout mice can be used to investigate the pathomechanisms of glaucoma more precisely.
Collapse
|
22
|
|
23
|
Alqawlaq S, Flanagan JG, Sivak JM. All roads lead to glaucoma: Induced retinal injury cascades contribute to a common neurodegenerative outcome. Exp Eye Res 2018; 183:88-97. [PMID: 30447198 DOI: 10.1016/j.exer.2018.11.005] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 11/02/2018] [Accepted: 11/06/2018] [Indexed: 11/25/2022]
Abstract
Glaucoma describes a distinct optic neuropathy with complex etiology and a variety of associated risk factors, but with similar pathological endpoints. Risk factors such as age, increased intraocular pressure (IOP), low mean arterial pressure, and autoimmune disease, can all be associated with death of retinal ganglion cells (RGCs) and optic nerve head remodeling. Today, IOP management remains the standard of care, even though IOP elevation is not pathognomonic of glaucoma, and patients can continue to lose vision despite effective IOP control. A contemporary view of glaucoma as a complex, neurodegenerative disease has developed, along with the recognition of a need for new disease modifying retinal treatment strategies and improved outcomes. However, the distinction between risk factors triggering the disease process and retinal injury responses is not always clear. In this review, we attempt to distinguish between the various triggers, and their association with subsequent key RGC injury mechanisms. We propose that distinct glaucomatous risk factors result in similar retinal and optic nerve injury cascades, including oxidative and metabolic stress, glial reactivity, and altered inflammatory responses, which induce common molecular signals to induce RGC apoptosis. This organization forms a coherent disease framework and presents conserved targets for therapeutic intervention that are not limited to specific risk factors.
Collapse
Affiliation(s)
- Samih Alqawlaq
- Department of Vision Science, Krembil Research Institute, University Health Network, Toronto, ON, Canada; Vision Science Research Program, Ophthalmology and Vision Science, University of Toronto, Toronto, ON, Canada; Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - John G Flanagan
- School of Optometry and Vision Science Program, University of California at Berkeley, Berkeley, CA, USA
| | - Jeremy M Sivak
- Department of Vision Science, Krembil Research Institute, University Health Network, Toronto, ON, Canada; Vision Science Research Program, Ophthalmology and Vision Science, University of Toronto, Toronto, ON, Canada; Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
24
|
Bell K, Und Hohenstein-Blaul NVT, Teister J, Grus F. Modulation of the Immune System for the Treatment of Glaucoma. Curr Neuropharmacol 2018; 16:942-958. [PMID: 28730968 PMCID: PMC6120111 DOI: 10.2174/1570159x15666170720094529] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 05/17/2017] [Accepted: 07/18/2017] [Indexed: 12/20/2022] Open
Abstract
Background: At present intraocular pressure (IOP) lowering therapies are the only approach to treat glaucoma. Neuroprotective strategies to protect the retinal ganglion cells (RGC) from apoptosis are lacking to date. Substantial amount of research concerning the role of the immune system in glaucoma has been performed in the recent years. This review aims to analyse changes found in the peripheral immune system, as well as selected local changes of retina immune cells in the glaucomatous retina. Methods: By dividing the immune system into the innate and the adaptive immune system, a systematic literature research was performed to find recent approaches concerning the modulation of the immune system in the context of glaucoma. Also ClinicalTrials.gov was assessed to identify studies with a translational context. Results: We found that some aspects of the immune system, such as changes in antibody levels, changes in toll like receptor signalling, T cells and retinal microglial cells, experience more research activity than other areas such as changes in dendritic cells or macrophages. Briefly, results from clinical studies revealed altered immunoreactivities against retinal and optic nerve antigens in sera and aqueous humor of glaucoma patients and point toward an autoimmune involvement in glaucomatous neurodegeneration and RGC death. IgG accumulations along with plasma cells were found localised in human glaucomatous retinae in a pro-inflammatory environment possibly maintained by microglia. Animal studies show that antibodies (e.g. anti- heat shock protein 60 and anti-myelin basic protein) elevated in glaucoma patients provoke autoaggressive RGC loss and are associated with IgG depositions and increased microglial cells. Also, studies addressing changes in T lymphocytes, macrophages but also local immune responses in the retina have been performed and also hold promising results. Conclusions: This recapitulation of recent literature demonstrates that the immune system definitely plays a role in the pathogenesis of glaucoma. Multiple changes in the peripheral innate as well as adaptive immune system have been detected and give room for further research concerning valuable therapeutic targets. We conclude that there still is a great need to bring together the results derived from basic research analysing different aspects of the immune system in glaucoma to understand the immune context of the disease. Furthermore local immune changes in the retina of glaucoma patients still leave room for further therapeutic targets
Collapse
Affiliation(s)
- Katharina Bell
- Experimental and Translational Ophthalmology Mainz, Department of Ophthalmology, Medical Center of the Johannes Gutenberg University Mainz; Langenbeckstrasse 1, 55101 Mainz, Germany
| | - Nadine von Thun Und Hohenstein-Blaul
- Experimental and Translational Ophthalmology Mainz, Department of Ophthalmology, Medical Center of the Johannes Gutenberg University Mainz; Langenbeckstrasse 1, 55101 Mainz, Germany
| | - Julia Teister
- Experimental and Translational Ophthalmology Mainz, Department of Ophthalmology, Medical Center of the Johannes Gutenberg University Mainz; Langenbeckstrasse 1, 55101 Mainz, Germany
| | - Franz Grus
- Experimental and Translational Ophthalmology Mainz, Department of Ophthalmology, Medical Center of the Johannes Gutenberg University Mainz; Langenbeckstrasse 1, 55101 Mainz, Germany
| |
Collapse
|
25
|
Kuehn S, Meißner W, Grotegut P, Theiss C, Dick HB, Joachim SC. Intravitreal S100B Injection Leads to Progressive Glaucoma Like Damage in Retina and Optic Nerve. Front Cell Neurosci 2018; 12:312. [PMID: 30319357 PMCID: PMC6169322 DOI: 10.3389/fncel.2018.00312] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 08/28/2018] [Indexed: 12/15/2022] Open
Abstract
The glial protein S100B, which belongs to a calcium binding protein family, is up-regulated in neurological diseases, like multiple sclerosis or glaucoma. In previous studies, S100B immunization led to retinal ganglion cell (RGC) loss in an experimental autoimmune glaucoma (EAG) model. Now, the direct degenerative impact of S100B on the retina and optic nerve was evaluated. Therefore, 2 μl of S100B was intravitreally injected in two concentrations (0.2 and 0.5 μg/μl). At day 3, 14 and 21, retinal neurons, such as RGCs, amacrine and bipolar cells, as well as apoptotic mechanisms were analyzed. Furthermore, neurofilaments, myelin fibers and axons of optic nerves were evaluated. In addition, retinal function and immunoglobulin G (IgG) level in the serum were measured. At day 3, RGCs were unaffected in the S100B groups, when compared to the PBS group. Later, at days 14 and 21, the RGC number as well as the β-III tubulin protein level was reduced in the S100B groups. Only at day 14, active apoptotic mechanisms were noted. The number of amacrine cells was first affected at day 21, while the bipolar cell amount remained comparable to the PBS group. Also, the optic nerve neurofilament structure was damaged from day 3 on. At day 14, numerous swollen axons were observed. The intraocular injection of S100B is a new model for a glaucoma like degeneration. Although the application site was the eye, the optic nerve degenerated first, already at day 3. From day 14 on, retinal damage and loss of function was noted. The RGCs in the middle part of the retina were first affected. At day 21, the damage expanded and RGCs had degenerated in all areas of the retina as well as amacrine cells. Furthermore, elevated IgG levels in the serum were measured at day 21, which could be a sign of a late and S100B independet immune response. In summary, S100B had a direct destroying impact on the axons of the optic nerve. The damage of the retinal cell bodies seems to be a consequence of this axon loss.
Collapse
Affiliation(s)
- Sandra Kuehn
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Wilhelm Meißner
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Pia Grotegut
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Carsten Theiss
- Department of Cytology, Institute of Anatomy, Ruhr-University Bochum, Bochum, Germany
| | - H Burkhard Dick
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Stephanie C Joachim
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| |
Collapse
|
26
|
S100B immunization triggers NFκB and complement activation in an autoimmune glaucoma model. Sci Rep 2018; 8:9821. [PMID: 29959432 PMCID: PMC6026137 DOI: 10.1038/s41598-018-28183-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 06/13/2018] [Indexed: 12/27/2022] Open
Abstract
In glaucoma, latest studies revealed an involvement of the complement system with and without an elevated intraocular pressure. In the experimental autoimmune glaucoma model, immunization with antigens, such as S100B, lead to retinal ganglion cell (RGC) loss and optic nerve degeneration after 28 days. Here, we investigated the timeline of progression of the complement system, toll-like-receptor 4 (TLR4), and the transcription factor nucleus factor-kappa B (NFκB). Therefore, rats were immunized with S100B protein (S100) and analyzed at 3, 7, and 14 days. RGC numbers were comparable at all points in time, whereas a destruction of S100 optic nerves was noted at 14 days. A significant increase of mannose binding lectin (MBL) was observed in S100 retinas at 3 days. Subsequently, significantly more MBL+ cells were seen in S100 optic nerves at 7 and 14 days. Accordingly, C3 was upregulated in S100 retinas at 14 days. An increase of interleukin-1 beta was noted in S100 aqueous humor samples at 7 days. In this study, activation of complement system via the lectin pathway was obvious. However, no TLR4 alterations were noted in S100 retinas and optic nerves. Interestingly, a significant NFκB increase was observed in S100 retinas at 7 and 14 days. We assume that NFκB activation might be triggered via MBL leading to glaucomatous damage.
Collapse
|
27
|
Wilmes AT, Reinehr S, Kühn S, Pedreiturria X, Petrikowski L, Faissner S, Ayzenberg I, Stute G, Gold R, Dick HB, Kleiter I, Joachim SC. Laquinimod protects the optic nerve and retina in an experimental autoimmune encephalomyelitis model. J Neuroinflammation 2018; 15:183. [PMID: 29903027 PMCID: PMC6002998 DOI: 10.1186/s12974-018-1208-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 05/20/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The oral immunomodulatory agent laquinimod is currently evaluated for multiple sclerosis (MS) treatment. Phase II and III studies demonstrated a reduction of degenerative processes. In addition to anti-inflammatory effects, laquinimod might have neuroprotective properties, but its impact on the visual system, which is often affected by MS, is unknown. The aim of our study was to investigate potential protective effects of laquinimod on the optic nerve and retina in an experimental autoimmune encephalomyelitis (EAE) model. METHODS We induced EAE in C57/BL6 mice via MOG35-55 immunization. Animals were divided into an untreated EAE group, three EAE groups receiving laquinimod (1, 5, or 25 mg/kg daily), starting the day post-immunization, and a non-immunized control group. Thirty days post-immunization, scotopic electroretinograms were carried out, and mice were sacrificed for histopathology (HE, LFB), immunohistochemistry (MBP, Iba1, Tmem119, F4/80, GFAP, vimentin, Brn-3a, cleaved caspase 3) of the optic nerve and retina, and retinal qRT-PCR analyses (Brn-3a, Iba1, Tmem119, AMWAP, CD68, GFAP). To evaluate the effect of a therapeutic approach, EAE animals were treated with 25 mg/kg laquinimod from day 16 when 60% of the animals had developed clinical signs of EAE. RESULTS Laquinimod reduced neurological EAE symptoms and improved the neuronal electrical output of the inner nuclear layer compared to untreated EAE mice. Furthermore, cellular infiltration, especially recruited phagocytes, and demyelination in the optic nerve were reduced. Microglia were diminished in optic nerve and retina. Retinal macroglial signal was reduced under treatment, whereas in the optic nerve macroglia were not affected. Additionally, laquinimod preserved retinal ganglion cells and reduced apoptosis. A later treatment with laquinimod in a therapeutic approach led to a reduction of clinical signs and to an improved b-wave amplitude. However, no changes in cellular infiltration and demyelination of the optic nerves were observed. Also, the number of retinal ganglion cells remained unaltered. CONCLUSION From our study, we deduce neuroprotective and anti-inflammatory effects of laquinimod on the optic nerve and retina in EAE mice, when animals were treated before any clinical signs were noted. Given the fact that the visual system is frequently affected by MS, the agent might be an interesting subject of further neuro-ophthalmic investigations.
Collapse
Affiliation(s)
- Anna T Wilmes
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, In der Schornau 23-25, 44892, Bochum, Germany
| | - Sabrina Reinehr
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, In der Schornau 23-25, 44892, Bochum, Germany
| | - Sandra Kühn
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, In der Schornau 23-25, 44892, Bochum, Germany
| | - Xiomara Pedreiturria
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, Gudrunstrasse 56, 44791, Bochum, Germany
| | - Laura Petrikowski
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, Gudrunstrasse 56, 44791, Bochum, Germany
| | - Simon Faissner
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, Gudrunstrasse 56, 44791, Bochum, Germany
| | - Ilya Ayzenberg
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, Gudrunstrasse 56, 44791, Bochum, Germany
| | - Gesa Stute
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, In der Schornau 23-25, 44892, Bochum, Germany
| | - Ralf Gold
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, Gudrunstrasse 56, 44791, Bochum, Germany
| | - H Burkhard Dick
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, In der Schornau 23-25, 44892, Bochum, Germany
| | - Ingo Kleiter
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, Gudrunstrasse 56, 44791, Bochum, Germany.
| | - Stephanie C Joachim
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, In der Schornau 23-25, 44892, Bochum, Germany.
| |
Collapse
|
28
|
Schmelter C, Perumal N, Funke S, Bell K, Pfeiffer N, Grus FH. Peptides of the variable IgG domain as potential biomarker candidates in primary open-angle glaucoma (POAG). Hum Mol Genet 2018; 26:4451-4464. [PMID: 29036575 DOI: 10.1093/hmg/ddx332] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 08/15/2017] [Indexed: 12/17/2022] Open
Abstract
Autoantibody profiling has gained increasing interest in the research field of glaucoma promising the detection of highly specific and sensitive marker candidates for future diagnostic purposes. Recent studies demonstrated that immune responses are characterized by the expression of congruent or similar complementarity determining regions (CDR) in different individuals and could be used as molecular targets in biomarker discovery. Main objective of this study was to characterize glaucoma-specific peptides from the variable region of sera-derived immunoglobulins using liquid chromatography--mass spectrometry (LC-MS)-based quantitative proteomics. IgG was purified from sera of 13 primary open-angle glaucoma patients (POAG) and 15 controls (CTRL) and subsequently digested into Fab and Fc by papain. Fab was further purified, tryptic digested and measured by LC-MS/MS. Discovery proteomics revealed in total 75 peptides of the variable IgG domain showing significant glaucoma-related level changes (P < 0.05; log2 fold change ≥ 0.5): 6 peptides were high abundant in POAG sera, whereas 69 peptides were low abundant in comparison to CTRL group. Via accurate inclusion mass screening strategy 28 IgG V domain peptides were further validated showing significantly decreased expression levels in POAG sera. Amongst others 5 CDR1, 2 CDR2 and 1 CDR3 sequences. In addition, we observed significant shifts in the variable heavy chain family distribution and disturbed κ/λ ratios in POAG patients in contrast to CTRL. These findings strongly indicate that glaucoma is accompanied by systemic effects on antibody production and B cell maturation possibly offering new prospects for future diagnostic or therapy purposes.
Collapse
Affiliation(s)
- Carsten Schmelter
- Department of Experimental and Translational Ophthalmology, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Natarajan Perumal
- Department of Experimental and Translational Ophthalmology, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Sebastian Funke
- Department of Experimental and Translational Ophthalmology, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Katharina Bell
- Department of Experimental and Translational Ophthalmology, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Norbert Pfeiffer
- Department of Experimental and Translational Ophthalmology, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Franz H Grus
- Department of Experimental and Translational Ophthalmology, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
29
|
Reinehr S, Kuehn S, Casola C, Koch D, Stute G, Grotegut P, Dick HB, Joachim SC. HSP27 immunization reinforces AII amacrine cell and synapse damage induced by S100 in an autoimmune glaucoma model. Cell Tissue Res 2017; 371:237-249. [PMID: 29064077 DOI: 10.1007/s00441-017-2710-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 10/11/2017] [Indexed: 12/11/2022]
Abstract
Previous studies have revealed a loss of retinal ganglion cells (RGCs) and optic nerve fibers after immunization with the S100B protein. Addition of heat shock protein 27 (HSP27) also leads to a decrease of RGCs. Our present aim has been to analyze various retinal cell types after immunization with S100B or S100B + HSP27 (S100 + HSP). After 28 days, retinas were processed for immunohistology and Western blot. RGCs, immunostained for NeuN, were significantly decreased in the S100 and the S100 + HSP groups. Significantly fewer ChAT+ cells were noted in both groups, whereas parvalbumin+ cells were only affected in the S100 + HSP group. Western blot results also revealed fewer ChAT signals in both immunized groups. No changes were noted with regard to PKCα+ rod bipolar cells, whereas a significant loss of recoverin+ cone bipolar cells was observed in both groups via immunohistology and Western blot. The presynaptic marker Bassoon and the postsynaptic marker PSD95 were significantly reduced in the S100 + HSP group. Opsin+ and rhodopsin+ photoreceptors revealed no changes in either group. Thus, the inner retinal layers are affected by immunization. However, the combination of S100 and HSP27 has a stronger additive effect on the retinal synapses and AII amacrine cells.
Collapse
Affiliation(s)
- Sabrina Reinehr
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, In der Schornau 23-25, 44892, Bochum, Germany
| | - Sandra Kuehn
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, In der Schornau 23-25, 44892, Bochum, Germany
| | - Christina Casola
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, In der Schornau 23-25, 44892, Bochum, Germany
| | - Dennis Koch
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, In der Schornau 23-25, 44892, Bochum, Germany
| | - Gesa Stute
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, In der Schornau 23-25, 44892, Bochum, Germany
| | - Pia Grotegut
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, In der Schornau 23-25, 44892, Bochum, Germany
| | - H Burkhard Dick
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, In der Schornau 23-25, 44892, Bochum, Germany
| | - Stephanie C Joachim
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, In der Schornau 23-25, 44892, Bochum, Germany.
| |
Collapse
|
30
|
Thomas CN, Berry M, Logan A, Blanch RJ, Ahmed Z. Caspases in retinal ganglion cell death and axon regeneration. Cell Death Discov 2017; 3:17032. [PMID: 29675270 PMCID: PMC5903394 DOI: 10.1038/cddiscovery.2017.32] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 03/31/2017] [Accepted: 04/23/2017] [Indexed: 02/07/2023] Open
Abstract
Retinal ganglion cells (RGC) are terminally differentiated CNS neurons that possess limited endogenous regenerative capacity after injury and thus RGC death causes permanent visual loss. RGC die by caspase-dependent mechanisms, including apoptosis, during development, after ocular injury and in progressive degenerative diseases of the eye and optic nerve, such as glaucoma, anterior ischemic optic neuropathy, diabetic retinopathy and multiple sclerosis. Inhibition of caspases through genetic or pharmacological approaches can arrest the apoptotic cascade and protect a proportion of RGC. Novel findings have also highlighted a pyroptotic role of inflammatory caspases in RGC death. In this review, we discuss the molecular signalling mechanisms of apoptotic and inflammatory caspase responses in RGC specifically, their involvement in RGC degeneration and explore their potential as therapeutic targets.
Collapse
Affiliation(s)
- Chloe N Thomas
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Martin Berry
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Ann Logan
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Richard J Blanch
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK.,Academic Department of Military Surgery and Trauma, Royal Centre for Defence Medicine, Birmingham, UK
| | - Zubair Ahmed
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| |
Collapse
|
31
|
Kuehn S, Hurst J, Jashari A, Ahrens K, Tsai T, Wunderlich IM, Dick HB, Joachim SC, Schnichels S. The novel induction of retinal ganglion cell apoptosis in porcine organ culture by NMDA - an opportunity for the replacement of animals in experiments. Altern Lab Anim 2017; 44:557-568. [PMID: 28094536 DOI: 10.1177/026119291604400608] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Some of the advantages of retina organ culture models include their efficient and easy handling and the ability to standardise relevant parameters. Additionally, when porcine eyes are obtained from the food industry, no animals are killed solely for research purposes. To induce retinal degeneration, a commonly used toxic substance, N-methyl-D-aspartate (NMDA), was applied to the cultures. To this end, organotypic cultures of porcine retinas were cultured and treated with different doses of NMDA (0 [control], 50, 100 and 200μM) on day 2 for 48 hours. On day 7, the retinas were cryo-conserved for histological, Western blot and quantitative rt-PCR (qrt-PCR) analyses. NMDA treatment was found to significantly increase retinal ganglion cell (RGC) apoptosis in all the treated groups, without a profound RGC loss. In addition, the intrinsic apoptotic pathway was activated in the 50μM and 100μM NMDA groups, whereas induced nitric oxide synthase (iNOS) expression was increased in the 200μM group. A slight microglial response was detectable, especially in the 100μM group. NMDA treatment induced apoptosis, oxidative stress and a slight microglia activation. All these effects mimic a chronic slow progressive disease that especially affects RGCs, such as glaucoma. A particular advantage of this model is that mediators that can interact in the very early stages of the onset of RGC death, can be easily detected and potential therapies can be tested.
Collapse
Affiliation(s)
- Sandra Kuehn
- Experimental Eye Research, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Jose Hurst
- University Eye Hospital Tübingen, Centre for Ophthalmology, Tübingen, Germany
| | - Adelina Jashari
- Experimental Eye Research, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Kathrin Ahrens
- Experimental Eye Research, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Teresa Tsai
- Experimental Eye Research, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Ilan M Wunderlich
- Experimental Eye Research, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | - H Burkhard Dick
- Experimental Eye Research, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Stephanie C Joachim
- Experimental Eye Research, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Sven Schnichels
- University Eye Hospital Tübingen, Centre for Ophthalmology, Tübingen, Germany
| |
Collapse
|
32
|
Hurst J, Kuehn S, Jashari A, Tsai T, Bartz-Schmidt KU, Schnichels S, Joachim SC. A novel porcine ex vivo retina culture model for oxidative stress induced by H₂O₂. Altern Lab Anim 2017; 45:11-25. [PMID: 28409994 DOI: 10.1177/026119291704500105] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Oxidative stress is a key player in many ophthalmic diseases. However, the role of oxidative stress in most degenerative processes is not yet known. Therefore, accurate and practical models are required to efficiently screen for therapeutics. Porcine eyes are closely related to the human eye, and can be obtained from the abattoir as a by-product of the food industry. Therefore, they offer excellent opportunities for the development of culture models with which to pre-screen potential therapies, while reducing the use of laboratory animals. To induce oxidative stress, organotypic cultures of porcine retina were treated with different doses of hydrogen peroxide (H₂O₂; 100, 300 and 500μM) for three hours. On days 3 and 8, the retinas were conserved for histological and Western blotting analyses and for evaluation of gene expression, which determined the number of retinal ganglion cells (RGCs), the activation state of glial cells, and the expression levels of several oxidative stress markers. H₂O₂ treatment led to a reduction in the number of RGCs and to an increase in apoptotic RGCs. In addition, a dose-dependent increase of microglia and an elevation of CD11b expression was observed. On day 3, a reduction of IL-1β, and an increase of iNOS, as well as of HSP70 mRNA were found. On day 8, an increase in TNF-α and IL-1β mRNA expression was detected. In conclusion, this ex vivo model offers an opportunity to study the molecular mechanisms underlying certain eye disorders and to test new therapeutic approaches to diminish the effects of oxidative stress.
Collapse
Affiliation(s)
- José Hurst
- University Eye Hospital Tübingen, Centre for Ophthalmology Tübingen, Tübingen, Germany
| | - Sandra Kuehn
- Experimental Eye Research, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Adelina Jashari
- Experimental Eye Research, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Teresa Tsai
- Experimental Eye Research, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | | | - Sven Schnichels
- University Eye Hospital Tübingen, Centre for Ophthalmology Tübingen, Tübingen, Germany
| | - Stephanie C Joachim
- Experimental Eye Research, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| |
Collapse
|
33
|
Lorenz K, Beck S, Keilani MM, Wasielica-Poslednik J, Pfeiffer N, Grus FH. Course of serum autoantibodies in patients after acute angle-closure glaucoma attack. Clin Exp Ophthalmol 2017; 45:280-287. [PMID: 27758063 DOI: 10.1111/ceo.12864] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 09/15/2016] [Accepted: 09/19/2016] [Indexed: 12/15/2022]
Abstract
BACKGROUND The aim of our investigation was to analyze the autoantibody -reactivities of patients after acute angle-closure glaucoma (AACG) by means of a protein microarray approach to identify intraocular pressure(IOP)-dependent antibodies. METHODS Collected sera from different study time points (AACG n = 6, 0, 2, 4 and 12 weeks) and control group (CTRL n = 11, 0 and 12 weeks) were analyzed. Protein-microarrays were incubated with sera, and occurring immunoreactivities were visualized with fluorescence labeled secondary antibodies. To detect changes, spot intensities were digitized and compared with statistical techniques. RESULTS Three autoantibodies with significant level-alteration in the time course of the survey could be identified. Immunoreactivities to heat shock 27-kDa protein (HSP27), tubulin-tyrosine ligase-like protein 12 (TTLL12), and neuron-specific enolase (NSE) show an increasing linear trend from week 0 up to week 12 with a positive correlation coefficient (P ≤ 0.05, r ≥ 0.4). In the CTRL- group, no significant alterations could be detected in corresponding autoantibody-level. Analysis of variance revealed significant changes of antibody-level between certain time points (anti-HSP27 antibody [week 0 vs. 2], anti-TTLL12 antibody [week 0 vs. 12], and anti-NSE antibody [week 4 vs. 12] [P ≤ 0.05, respectively]) in AACG group. CONCLUSIONS With this autoantibodies profiling approach, we were able to detect autoimmune reactivities in sera of patients without former indication for glaucomatous damage after rise of IOP due to AACG attack. After further validation in subsequent studies, this autoantibodies could give further insights into the pathogenesis of glaucoma and could possibly help to understand the effect of IOP on glaucomatous optic neuropathy.
Collapse
Affiliation(s)
- Katrin Lorenz
- Department of Experimental Ophthalmology, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Sabine Beck
- Department of Experimental Ophthalmology, University Medical Center, Johannes Gutenberg-University, Mainz, Germany.,Department of Pharmacy and Biochemistry, Johannes Gutenberg-University Mainz, Germany
| | - Munir M Keilani
- Department of Experimental Ophthalmology, University Medical Center, Johannes Gutenberg-University, Mainz, Germany.,Vision 100 Die Augenärzte, Gemeinschaftspraxis Mönchengladbach, Germany
| | - Joanna Wasielica-Poslednik
- Department of Experimental Ophthalmology, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Norbert Pfeiffer
- Department of Experimental Ophthalmology, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Franz H Grus
- Department of Experimental Ophthalmology, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| |
Collapse
|
34
|
Von Thun Und Hohenstein-Blaul N, Kunst S, Pfeiffer N, Grus FH. Biomarkers for glaucoma: from the lab to the clinic. Eye (Lond) 2017; 31:225-231. [PMID: 28085137 DOI: 10.1038/eye.2016.300] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 11/11/2016] [Indexed: 11/09/2022] Open
Abstract
Glaucoma, a leading cause of irreversible blindness worldwide, is often not diagnosed until many years after disease onset. Early and objective diagnostic measures are yet missing. Besides the main risk factor, an elevated intraocular pressure (IOP), age, sex, and ethnicity are known to affect disease progression and severity. Furthermore, oxidative stress, elevated glutamate concentrations, and an autoimmune component are considered possible risk factors. We could identify several potential proteomic biomarkers in glaucoma and examine distinct changes in the glaucomatous human retina proteome. Using an experimental autoimmune glaucoma animal (EAG) model we could demonstrate an IOP-independent loss of retinal ganglion cells (RGC), which is accompanied by antibody depositions and increased levels of microglia. In a different animal model we showed that intermittent IOP elevations provoke neurodegeneration in the optic nerve and the retina and elicit changes of IgG autoantibody reactivities. The correlation between neuronal damage and changes in autoantibody reactivity suggests that autoantibody profiling could be a useful biomarker for glaucoma. In vivo studies on neuroretinal cells and porcine retinal explants demonstrated a protective effect of antibodies (eg, anti-GFAP) on RGC, which seems to be the result of reduced stress levels in the retina. We conclude that the absence of some autoantibodies in glaucoma patients reflects a loss of the protective potential of natural autoimmunity and may thus encourage neurodegenerative processes. Concluding, autoantibody profiles resemble useful biomarkers for diagnosis, progression and severity of glaucoma. Future longitudinal studies will help to improve early detection and enable better monitoring of disease progression.
Collapse
Affiliation(s)
- N Von Thun Und Hohenstein-Blaul
- Experimental and Translational Ophthalmology, Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - S Kunst
- Experimental and Translational Ophthalmology, Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - N Pfeiffer
- Experimental and Translational Ophthalmology, Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - F H Grus
- Experimental and Translational Ophthalmology, Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
35
|
Kuehn S, Hurst J, Rensinghoff F, Tsai T, Grauthoff S, Satgunarajah Y, Dick HB, Schnichels S, Joachim SC. Degenerative effects of cobalt-chloride treatment on neurons and microglia in a porcine retina organ culture model. Exp Eye Res 2017; 155:107-120. [PMID: 28089775 DOI: 10.1016/j.exer.2017.01.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 12/20/2016] [Accepted: 01/11/2017] [Indexed: 01/08/2023]
Abstract
In order to understand the pathological processes of retinal diseases, experimental models are necessary. Cobalt, as part of the vitamin B12 complex, is important for neuronal integrity. However, it is known that high quantities of cobalt induce cytotoxic mechanisms via hypoxia mimicry. Therefore, we tested the degenerative effect of cobalt chloride (CoCl2) on neurons and microglia in a porcine retina organ culture model. Organotypic cultures of porcine retinas were cultured and treated with different concentrations of CoCl2 (0, 100, 300 and 500 μM) for 48 h. After four and eight days, CoCl2 induced a strong degeneration of the porcine retina, starting at 300 μM. A loss of retinal ganglion cells (RGCs, Brn-3a), amacrine cells (calretinin) and bipolar cells (PKCα) was observed. Additionally, a high expression of hypoxia induced factor-1a (HIF-1a) and heat shock protein 70 (HSP70) was noted at both points in time. Also, the Caspase 3 protein was activated and P21 expression was induced. However, only at day four, the Bax/Bcl-2 ratio was increased. The effect of CoCl2 was not restricted to neurons. CoCl2 concentrations reduced the microglia amount (Iba1) and activity (Iba1 + Fcγ-Receptor) at both points in time. These damaging effects on microglia were surprising, since CoCl2 causes hypoxia and a pro-inflammatory environment. However, high concentrations of CoCl2 also seem to be toxic to these cells. Similar degenerative mechanisms as in comparison to retinal ischemia animal models were observed. In summary, an effective and reproducible hypoxia-mimicking organotypic model for retinal degeneration was established, which is easy to handle and ready for drug studies.
Collapse
Affiliation(s)
- S Kuehn
- Experimental Eye Research, Eye Hospital, Ruhr-University Bochum, In der Schornau 23-25, 44892 Bochum, Germany
| | - J Hurst
- University Eye Hospital Tübingen, Centre for Ophthalmology, Elfriede-Aulhorn-Str. 7, 72076, Tübingen, Germany
| | - F Rensinghoff
- Experimental Eye Research, Eye Hospital, Ruhr-University Bochum, In der Schornau 23-25, 44892 Bochum, Germany
| | - T Tsai
- Experimental Eye Research, Eye Hospital, Ruhr-University Bochum, In der Schornau 23-25, 44892 Bochum, Germany
| | - S Grauthoff
- Experimental Eye Research, Eye Hospital, Ruhr-University Bochum, In der Schornau 23-25, 44892 Bochum, Germany
| | - Y Satgunarajah
- Experimental Eye Research, Eye Hospital, Ruhr-University Bochum, In der Schornau 23-25, 44892 Bochum, Germany
| | - H B Dick
- Experimental Eye Research, Eye Hospital, Ruhr-University Bochum, In der Schornau 23-25, 44892 Bochum, Germany
| | - S Schnichels
- University Eye Hospital Tübingen, Centre for Ophthalmology, Elfriede-Aulhorn-Str. 7, 72076, Tübingen, Germany.
| | - S C Joachim
- Experimental Eye Research, Eye Hospital, Ruhr-University Bochum, In der Schornau 23-25, 44892 Bochum, Germany.
| |
Collapse
|
36
|
Longitudinal Analysis of Serum Autoantibody-Reactivities in Patients with Primary Open Angle Glaucoma and Optic Disc Hemorrhage. PLoS One 2016; 11:e0166813. [PMID: 28030545 PMCID: PMC5193360 DOI: 10.1371/journal.pone.0166813] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 11/04/2016] [Indexed: 11/06/2022] Open
Abstract
Background The aim of our current investigation was to analyze the autoantibody-reactivities of primary open angle glaucoma patients with optic disc hemorrhage as possibly correlated to disease progression by means of a protein microarray approach. Methods Sera of patients with primary open angle glaucoma and optic disc hemorrhage (n = 16) were collected directly after study inclusion (0 weeks) and after 2 weeks, 4 weeks and 12 weeks. As a control group patients with primary open angle glaucoma (n = 18) were used (0 weeks and 12 weeks). Microarrays were incubated and occurring antibody-antigen-reactions were visualized with fluorescence labeled anti-human-IgG secondary antibodies. To detect changes in autoantibodies spot intensities were digitized and compared. Results With respect to the immunoreactivity at 0 weeks level increment of anti-adaptor protein 1 complex subunit mu-1 antibodies and anti-SPRY domain-containing SOCS box protein 3 antibodies in sera of primary open angle patients with optic disc hemorrhage was detected. Linear trend analysis revealed a positive correlation with r ≥ 0.8 between antibody-level and time course. Control group show no relevant changes in the same period. Significant changes were found in time point 4 comparison between patient groups in anti-adaptor protein 1 complex subunit mu-1-level (p = 0.01). No significant changes in visual acuity were found. Conclusion With this approach we were able to detect autoimmune reactivities in sera of patients with primary open angle glaucoma and optic disc hemorrhage compared to patients without optic disc hemorrhage. These antibodies could give further insights into the pathogenesis and the autoimmune component of glaucomatous optic neuropathy.
Collapse
|
37
|
Abstract
The term glaucoma summarizes a group of eye diseases that are accompanied by impairments of the optic nerve and related visual field deficits. An early diagnosis of glaucoma is currently not possible due to a lack of diagnostic tests; therefore, in most cases the disease is diagnosed many years after onset, which prevents an early therapy. The known risk factors for the development and progression of glaucomatous optic neuropathy comprise elevated intraocular pressure and a broad range of pressure fluctuations as well as lipometabolic disorders, genetic factor and diabetes. The consequences include the induction of anti-inflammatory proteins, elevated levels of oxidative stress and the destruction of retinal ganglion cells. Changes in the autoantibody repertoire have also been observed in the course of the disease. Basic ophthalmological research therefore focuses on the investigation of basic biochemical processes in the course of the disease. A better understanding of physiological and biochemical events is sought in order to develop new and more sensitive diagnostic options and to allow more targeted therapeutic measures. The understanding of biochemical processes allows a better insight into glaucoma progression to be gained, which will lead to improvements in diagnosis and therapy.
Collapse
|
38
|
Reinehr S, Reinhard J, Wiemann S, Stute G, Kuehn S, Woestmann J, Dick HB, Faissner A, Joachim SC. Early remodelling of the extracellular matrix proteins tenascin-C and phosphacan in retina and optic nerve of an experimental autoimmune glaucoma model. J Cell Mol Med 2016; 20:2122-2137. [PMID: 27374750 PMCID: PMC5082392 DOI: 10.1111/jcmm.12909] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 04/19/2016] [Indexed: 12/17/2022] Open
Abstract
Glaucoma is characterized by the loss of retinal ganglion cells (RGCs) and optic nerve fibres. Previous studies noted fewer RGCs after immunization with ocular antigens at 28 days. It is known that changes in extracellular matrix (ECM) components conduct retina and optic nerve degeneration. Here, we focused on the remodelling of tenascin‐C and phosphacan/receptor protein tyrosine phosphatase β/ζ in an autoimmune glaucoma model. Rats were immunized with optic nerve homogenate (ONA) or S100B protein (S100). Controls received sodium chloride (Co). After 14 days, no changes in RGC number were noted in all groups. An increase in GFAPmRNA expression was observed in the S100 group, whereas no alterations were noted via immunohistochemistry in both groups. Extracellular matrix remodelling was analyzed after 3, 7, 14 and 28 days. Tenascin‐C and 473HD immunoreactivity in retinae and optic nerves was unaltered in both immunized groups at 3 days. At 7 days, tenascin‐C staining increased in both tissues in the ONA group. Also, in the optic nerves of the S100 group, an intense tenascin‐C staining could be shown. In the retina, an increased tenascin‐C expression was also observed in ONA animals via Western blot. 473HD immunoreactivity was elevated in the ONA group in both tissues and in the S100 optic nerves at 7 days. At 14 days, tenascin‐C and 473HD immunoreactivity was up‐regulated in the ONA retinae, whereas phosphacan expression was up‐regulated in both groups. We conclude that remodelling of tenascin‐C and phosphacan occurred shortly after immunization, already before RGC loss. We assume that both ECM molecules represent early indicators of neurodegeneration.
Collapse
Affiliation(s)
- Sabrina Reinehr
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Jacqueline Reinhard
- Department of Cell Morphology and Molecular Neurobiology, Ruhr-University Bochum, Bochum, Germany
| | - Susanne Wiemann
- Department of Cell Morphology and Molecular Neurobiology, Ruhr-University Bochum, Bochum, Germany
| | - Gesa Stute
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Sandra Kuehn
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Julia Woestmann
- Department of Cell Morphology and Molecular Neurobiology, Ruhr-University Bochum, Bochum, Germany
| | - H Burkhard Dick
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Andreas Faissner
- Department of Cell Morphology and Molecular Neurobiology, Ruhr-University Bochum, Bochum, Germany.
| | - Stephanie C Joachim
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany.
| |
Collapse
|
39
|
Reinehr S, Reinhard J, Gandej M, Kuehn S, Noristani R, Faissner A, Dick HB, Joachim SC. Simultaneous Complement Response via Lectin Pathway in Retina and Optic Nerve in an Experimental Autoimmune Glaucoma Model. Front Cell Neurosci 2016; 10:140. [PMID: 27313510 PMCID: PMC4887475 DOI: 10.3389/fncel.2016.00140] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 05/10/2016] [Indexed: 12/18/2022] Open
Abstract
Glaucoma is a multifactorial disease and especially mechanisms occurring independently from an elevated intraocular pressure (IOP) are still unknown. Likely, the immune system contributes to the glaucoma pathogenesis. Previously, IgG antibody depositions and retinal ganglion cell (RGC) loss were found in an IOP-independent autoimmune glaucoma model. Therefore, we investigated the possible participation of the complement system in this model. Here, rats were immunized with bovine optic nerve homogenate antigen (ONA), while controls (Co) received sodium chloride (n = 5–6/group). After 14 days, RGC density was quantified on flatmounts. No changes in the number of RGCs could be observed at this point in time. Longitudinal optic nerve sections were stained against the myelin basic protein (MBP). We could note few signs of degeneration processes. In order to detect distinct complement components, retinas and optic nerves were labeled with complement markers at 3, 7, 14, and 28 days and analyzed. Significantly more C3 and MAC depositions were found in retinas and optic nerves of the ONA group. These were already present at day 7, before RGC loss and demyelination occurred. Additionally, an upregulation of C3 protein was noted via Western Blot at this time. After 14 days, quantitative real-time PCR revealed significantly more C3 mRNA in the ONA retinas. An upregulation of the lectin pathway-associated mannose-serine-protease-2 (MASP2) was observed in the retinas as well as in the optic nerves of the ONA group after 7 days. Significantly more MASP2 in retinas could also be observed via Western Blot analyses at this point in time. No effect was noted in regard to C1q. Therefore, we assume that the immunization led to an activation of the complement system via the lectin pathway in retinas and optic nerves at an early stage in this glaucoma model. This activation seems to be an early response, which then triggers degeneration. These findings can help to develop novel therapy strategies for glaucoma patients.
Collapse
Affiliation(s)
- Sabrina Reinehr
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum Bochum, Germany
| | - Jacqueline Reinhard
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr-University Bochum Bochum, Germany
| | - Marcel Gandej
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum Bochum, Germany
| | - Sandra Kuehn
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum Bochum, Germany
| | - Rozina Noristani
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum Bochum, Germany
| | - Andreas Faissner
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr-University Bochum Bochum, Germany
| | - H Burkhard Dick
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum Bochum, Germany
| | - Stephanie C Joachim
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum Bochum, Germany
| |
Collapse
|
40
|
|
41
|
Gramlich OW, Teister J, Neumann M, Tao X, Beck S, von Pein HD, Pfeiffer N, Grus FH. Immune response after intermittent minimally invasive intraocular pressure elevations in an experimental animal model of glaucoma. J Neuroinflammation 2016; 13:82. [PMID: 27090083 PMCID: PMC4836145 DOI: 10.1186/s12974-016-0542-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 04/07/2016] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Elevated intraocular pressure (IOP), as well as fluctuations in IOP, is a main risk factor for glaucoma, but its pathogenic effect has not yet been clarified. Beyond the multifactorial pathology of the disease, autoimmune mechanisms seem to be linked to retinal ganglion cell (RGC) death. This study aimed to identify if intermittent IOP elevations in vivo (i) elicit neurodegeneration, (ii) provokes an immune response and (iii) whether progression of RGC loss can be attenuated by the B lymphocyte inhibitor Belimumab. METHODS Using an intermittent ocular hypertension model (iOHT), Long Evans rats (n = 21) underwent 27 unilateral simulations of a fluctuating pressure profile. Nine of these animals received Belimumab, and additional seven rats served as normotensive controls. Axonal density was analyzed in PPD-stained optic nerve cross-sections. Retinal cross-sections were immunostained against Brn3a, Iba1, and IgG autoantibody depositions. Serum IgG concentration and IgG reactivities were determined using ELISA and protein microarrays. Data was analyzed using ANOVA and Tukey HSD test (unequal N) or student's independent t test by groups. RESULTS A wavelike IOP profile led to a significant neurodegeneration of optic nerve axons (-10.6 %, p < 0.001) and RGC (-19.5 %, p = 0.02) in iOHT eyes compared with fellow eyes. Belimumab-treated animals only showed slightly higher axonal survival and reduced serum IgG concentration (-29 %) after iOHT. Neuroinflammatory events, indicated by significantly upregulated microglia activation and IgG autoantibody depositions, were shown in all injured retinas. Significantly elevated serum autoantibody immunoreactivities against glutathione-S-transferase, spectrin, and transferrin were observed after iOHT and were negatively correlated to the axon density. CONCLUSIONS Intermittent IOP elevations are sufficient to provoke neurodegeneration in the optic nerve and the retina and elicit changes of IgG autoantibody reactivities. Although the inhibition of B lymphocyte activation failed to ameliorate axonal survival, the correlation between damage and changes in the autoantibody reactivity suggests that autoantibody profiling could be useful as a biomarker for glaucoma.
Collapse
Affiliation(s)
- Oliver W Gramlich
- Experimental Ophthalmology, Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany.,Glaucoma Cell Biology Laboratory, Department of Ophthalmology and Visual Sciences, The University of Iowa, Iowa City, IA, 62242, USA
| | - Julia Teister
- Experimental Ophthalmology, Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Mareike Neumann
- Experimental Ophthalmology, Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Xue Tao
- Experimental Ophthalmology, Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Sabine Beck
- Experimental Ophthalmology, Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Harald D von Pein
- Department of Neuropathology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, Mainz, 55131, Germany
| | - Norbert Pfeiffer
- Experimental Ophthalmology, Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Franz H Grus
- Experimental Ophthalmology, Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany.
| |
Collapse
|
42
|
Autoimmune aspects in glaucoma. Eur J Pharmacol 2016; 787:105-18. [PMID: 27090926 DOI: 10.1016/j.ejphar.2016.04.031] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 03/31/2016] [Accepted: 04/14/2016] [Indexed: 12/27/2022]
Abstract
The pathogenesis of glaucoma, a common neurodegenerative disease, involves an immunologic component. Studies demonstrate changes of autoantibody concentrations against retinal and optic nerve head antigens in glaucoma patients. Furthermore we found antibody deposits in human glaucomatous retinae in a pro-inflammatory environment. Clinical studies showed up regulated, but also significantly down-regulated autoantibody levels. These antibodies belong to the natural autoimmunity. The upregulation of autoantibodies can be associated with fatal conditions, but several studies demonstrate that natural autoantibodies entail also neuroprotective characteristics and influence the protein expression of neuroretinal cells. A misbalance in the physiological equilibrium may shift from regulatory immunity into a neuroinflammatory degenerative process, what may lead to a predisposition to glaucoma. However, the protective nature of autoantibodies and the molecular mechanisms underlying the very sensitive equilibrium of natural autoimmunity between autoaggression and neuroprotection offer promising target sites for new therapeutic approaches. Finally, the changes in antibody profiles represent a new opportunity as highly sensitive and specific biomarkers for diagnostics purposes.
Collapse
|
43
|
Systemic ocular antigen immunization leads only to a minor secondary immune response. J Neuroimmunol 2016; 293:114-122. [DOI: 10.1016/j.jneuroim.2016.02.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 02/24/2016] [Accepted: 02/26/2016] [Indexed: 11/13/2022]
|
44
|
Retinal and Optic Nerve Damage is Associated with Early Glial Responses in an Experimental Autoimmune Glaucoma Model. J Mol Neurosci 2016; 58:470-82. [PMID: 26746422 DOI: 10.1007/s12031-015-0707-2] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 12/22/2015] [Indexed: 02/06/2023]
Abstract
It is well established that the immunization with ocular antigens causes a retinal ganglion cell (RGC) decline, which is accompanied by glia alterations. In this study, the degenerative effects of the immunization with an optic nerve homogenate (ONA) and its purified compound S100 were analyzed on retinas and optic nerves. Since a participation of glia cells in cell death mechanisms is currently discussed, rats were immunized with S100 or ONA. At 14 and 28 days, immune-histological and Western blot analyses were performed to investigate the optic nerve structure (SMI-32), retinal ganglion cells (Brn-3a), apoptosis (cleaved caspase 3, FasL), and glial profile (Iba1, ED1, GFAP, vimentin). Neurofilament dissolution in S100 animals was evident at 14 days (p = 0.047) and increased at 28 days (p = 0.01). ONA optic nerves remained intact at early stages and degenerated later on (p = 0.002). In both groups, RGC loss was detected via immune-histology and Western blot at 28 days (ONA: p = 0.02; S100: p = 0.005). Additionally, more Iba1(+) retinal microglia could be detected at early stages (ONA: p = 0.006; S100: p = 0.028). A slight astrocyte response was detected on Western blots only on ONA retinas (p = 0.01). Hence, the RGC and optic nerve decline was partly antigen dependent, while neuronal loss is paralleled by an early microglial response.
Collapse
|
45
|
Gramlich OW, Ding QJ, Zhu W, Cook A, Anderson MG, Kuehn MH. Adoptive transfer of immune cells from glaucomatous mice provokes retinal ganglion cell loss in recipients. Acta Neuropathol Commun 2015; 3:56. [PMID: 26374513 PMCID: PMC4591529 DOI: 10.1186/s40478-015-0234-y] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 08/21/2015] [Indexed: 12/02/2022] Open
Abstract
INTRODUCTION Several studies have indicated that autoimmune and neuroinflammatory processes contribute to the neurodegeneration of retinal ganglion cells in human glaucoma patients and in animal models. To test the involvement of cellular immune processes in the pathophysiology of retinal ganglion cell degeneration in vivo, we carried out adoptive transfer experiments from two independent genetic mouse models of glaucoma into normal recipient mice. RESULTS Our findings indicate that transfer results in a progressive loss of retinal ganglion cells and their axons despite normal intraocular pressure in recipient mice. Signs of pan-retinal inflammation were not detected. Similar findings were obtained following transfer of isolated T-lymphocytes, but not after transfer of splenocytes from immune deficient glaucomatous mice. Transferred lymphocytes were detected integrated in the spleen and in the retinal ganglion cell layer of recipient animals, albeit at very low frequencies. Furthermore, we observed cell-cell interaction between transferred T-cells and recipient microglia along with focal microglial activation in recipient eyes. CONCLUSION This study demonstrates that the pathophysiology of glaucomatous degeneration in the tested animal models includes T-cell mediated events that are capable of causing loss of healthy retinal ganglion cells.
Collapse
Affiliation(s)
- Oliver W Gramlich
- Department of Ophthalmology and Visual Sciences, The University of Iowa, Iowa City, 52242, IA, USA
- Center for the Prevention and Treatment of Visual Loss, Iowa City VA Health Care System, 3135C MERF, 375 Newton Road, Iowa City, IA, 52242, USA
| | - Qiong J Ding
- Department of Ophthalmology and Visual Sciences, The University of Iowa, Iowa City, 52242, IA, USA
| | - Wei Zhu
- Department of Ophthalmology and Visual Sciences, The University of Iowa, Iowa City, 52242, IA, USA
| | - Amy Cook
- Department of Ophthalmology and Visual Sciences, The University of Iowa, Iowa City, 52242, IA, USA
| | - Michael G Anderson
- Department of Ophthalmology and Visual Sciences, The University of Iowa, Iowa City, 52242, IA, USA
- Center for the Prevention and Treatment of Visual Loss, Iowa City VA Health Care System, 3135C MERF, 375 Newton Road, Iowa City, IA, 52242, USA
- Department of Molecular Physiology and Biophysics, The University of Iowa, Iowa City, 52242, IA, USA
| | - Markus H Kuehn
- Department of Ophthalmology and Visual Sciences, The University of Iowa, Iowa City, 52242, IA, USA.
- Center for the Prevention and Treatment of Visual Loss, Iowa City VA Health Care System, 3135C MERF, 375 Newton Road, Iowa City, IA, 52242, USA.
| |
Collapse
|
46
|
Madeira MH, Elvas F, Boia R, Gonçalves FQ, Cunha RA, Ambrósio AF, Santiago AR. Adenosine A2AR blockade prevents neuroinflammation-induced death of retinal ganglion cells caused by elevated pressure. J Neuroinflammation 2015; 12:115. [PMID: 26054642 PMCID: PMC4465153 DOI: 10.1186/s12974-015-0333-5] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 05/27/2015] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Elevated intraocular pressure (IOP) is a major risk factor for glaucoma, a degenerative disease characterized by the loss of retinal ganglion cells (RGCs). There is clinical and experimental evidence that neuroinflammation is involved in the pathogenesis of glaucoma. Since the blockade of adenosine A2A receptor (A2AR) confers robust neuroprotection and controls microglia reactivity in the brain, we now investigated the ability of A2AR blockade to control the reactivity of microglia and neuroinflammation as well as RGC loss in retinal organotypic cultures exposed to elevated hydrostatic pressure (EHP) or lipopolysaccharide (LPS). METHODS Retinal organotypic cultures were either incubated with LPS (3 μg/mL), to elicit a pro-inflammatory response, or exposed to EHP (+70 mmHg), to mimic increased IOP, for 4 or 24 h, in the presence or absence of the A2AR antagonist SCH 58261 (50 nM). A2AR expression, microglial reactivity and neuroinflammatory response were evaluated by immunohistochemistry, quantitative PCR (qPCR) and enzyme-linked immunosorbent assay (ELISA). RGC loss was assessed by immunohistochemistry. In order to investigate the contribution of pro-inflammatory mediators to RGC loss, the organotypic retinal cultures were incubated with rabbit anti-tumour necrosis factor (TNF) (2 μg/mL) and goat anti-interleukin-1β (IL-1β) (1 μg/mL) antibodies. RESULTS We report that the A2AR antagonist (SCH 58261) prevented microglia reactivity, increase in pro-inflammatory mediators as well as RGC loss upon exposure to either LPS or EHP. Additionally, neutralization of TNF and IL-1β prevented RGC loss induced by LPS or EHP. CONCLUSIONS This work demonstrates that A2AR blockade confers neuroprotection to RGCs by controlling microglia-mediated retinal neuroinflammation and prompts the hypothesis that A2AR antagonists may be a novel therapeutic option to manage glaucomatous disorders.
Collapse
Affiliation(s)
- Maria H Madeira
- Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, 3004-548, Coimbra, Portugal. .,CNC.IBILI, University of Coimbra, 3004-517, Coimbra, Portugal.
| | - Filipe Elvas
- Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, 3004-548, Coimbra, Portugal.
| | - Raquel Boia
- Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, 3004-548, Coimbra, Portugal. .,CNC.IBILI, University of Coimbra, 3004-517, Coimbra, Portugal.
| | - Francisco Q Gonçalves
- CNC.IBILI, University of Coimbra, 3004-517, Coimbra, Portugal. .,CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517, Coimbra, Portugal.
| | - Rodrigo A Cunha
- CNC.IBILI, University of Coimbra, 3004-517, Coimbra, Portugal. .,CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517, Coimbra, Portugal. .,Faculty of Medicine, University of Coimbra, 3000-548, Coimbra, Portugal.
| | - António Francisco Ambrósio
- Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, 3004-548, Coimbra, Portugal. .,CNC.IBILI, University of Coimbra, 3004-517, Coimbra, Portugal. .,Association for Innovation and Biomedical Research on Light (AIBILI), 3000-548, Coimbra, Portugal.
| | - Ana Raquel Santiago
- Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, 3004-548, Coimbra, Portugal. .,CNC.IBILI, University of Coimbra, 3004-517, Coimbra, Portugal. .,Association for Innovation and Biomedical Research on Light (AIBILI), 3000-548, Coimbra, Portugal. .,Faculty of Medicine, University of Coimbra, 3000-548, Coimbra, Portugal. .,IBILI, Faculty of Medicine, University of Coimbra, Azinhaga de Santa Comba, 3004-548, Coimbra, Portugal.
| |
Collapse
|
47
|
Madeira MH, Boia R, Santos PF, Ambrósio AF, Santiago AR. Contribution of microglia-mediated neuroinflammation to retinal degenerative diseases. Mediators Inflamm 2015; 2015:673090. [PMID: 25873768 PMCID: PMC4385698 DOI: 10.1155/2015/673090] [Citation(s) in RCA: 168] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 12/16/2014] [Indexed: 12/27/2022] Open
Abstract
Retinal degenerative diseases are major causes of vision loss and blindness worldwide and are characterized by chronic and progressive neuronal loss. One common feature of retinal degenerative diseases and brain neurodegenerative diseases is chronic neuroinflammation. There is growing evidence that retinal microglia, as in the brain, become activated in the course of retinal degenerative diseases, having a pivotal role in the initiation and propagation of the neurodegenerative process. A better understanding of the events elicited and mediated by retinal microglia will contribute to the clarification of disease etiology and might open new avenues for potential therapeutic interventions. This review aims at giving an overview of the roles of microglia-mediated neuroinflammation in major retinal degenerative diseases like glaucoma, age-related macular degeneration, and diabetic retinopathy.
Collapse
Affiliation(s)
- Maria H. Madeira
- Centre of Ophthalmology and Vision Sciences, IBILI, Faculty of Medicine, University of Coimbra, Azinhaga de Santa Comba, 3004-548 Coimbra, Portugal
| | - Raquel Boia
- Centre of Ophthalmology and Vision Sciences, IBILI, Faculty of Medicine, University of Coimbra, Azinhaga de Santa Comba, 3004-548 Coimbra, Portugal
| | - Paulo F. Santos
- Centre of Ophthalmology and Vision Sciences, IBILI, Faculty of Medicine, University of Coimbra, Azinhaga de Santa Comba, 3004-548 Coimbra, Portugal
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Department of Life Sciences, University of Coimbra, Coimbra, Portugal
| | - António F. Ambrósio
- Centre of Ophthalmology and Vision Sciences, IBILI, Faculty of Medicine, University of Coimbra, Azinhaga de Santa Comba, 3004-548 Coimbra, Portugal
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- AIBILI, Coimbra, Portugal
| | - Ana R. Santiago
- Centre of Ophthalmology and Vision Sciences, IBILI, Faculty of Medicine, University of Coimbra, Azinhaga de Santa Comba, 3004-548 Coimbra, Portugal
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- AIBILI, Coimbra, Portugal
| |
Collapse
|
48
|
|
49
|
Casola C, Schiwek JE, Reinehr S, Kuehn S, Grus FH, Kramer M, Dick HB, Joachim SC. S100 Alone Has the Same Destructive Effect on Retinal Ganglion Cells as in Combination with HSP 27 in an Autoimmune Glaucoma Model. J Mol Neurosci 2015; 56:228-36. [DOI: 10.1007/s12031-014-0485-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 12/21/2014] [Indexed: 01/27/2023]
|
50
|
|