51
|
Cell-Type-Specific Complement Profiling in the ABCA4 -/- Mouse Model of Stargardt Disease. Int J Mol Sci 2020; 21:ijms21228468. [PMID: 33187113 PMCID: PMC7697683 DOI: 10.3390/ijms21228468] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/02/2020] [Accepted: 11/09/2020] [Indexed: 12/13/2022] Open
Abstract
Stargardt macular degeneration is an inherited retinal disease caused by mutations in the ATP-binding cassette subfamily A member 4 (ABCA4) gene. Here, we characterized the complement expression profile in ABCA4−/− retinae and aligned these findings with morphological markers of retinal degeneration. We found an enhanced retinal pigment epithelium (RPE) autofluorescence, cell loss in the inner retina of ABCA4−/− mice and demonstrated age-related differences in complement expression in various retinal cell types irrespective of the genotype. However, 24-week-old ABCA4−/− mice expressed more c3 in the RPE and fewer cfi transcripts in the microglia compared to controls. At the protein level, the decrease of complement inhibitors (complement factor I, CFI) in retinae, as well as an increased C3b/C3 ratio in the RPE/choroid and retinae of ABCA4−/−, mice was confirmed. We showed a corresponding increase of the C3d/C3 ratio in the serum of ABCA4−/− mice, while no changes were observed for CFI. Our findings suggest an overactive complement cascade in the ABCA4−/− retinae that possibly contributes to pathological alterations, including microglial activation and neurodegeneration. Overall, this underpins the importance of well-balanced complement homeostasis to maintain retinal integrity.
Collapse
|
52
|
Ortega JT, Parmar T, Golczak M, Jastrzebska B. Protective Effects of Flavonoids in Acute Models of Light-Induced Retinal Degeneration. Mol Pharmacol 2020; 99:60-77. [PMID: 33154094 DOI: 10.1124/molpharm.120.000072] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 10/26/2020] [Indexed: 12/17/2022] Open
Abstract
Degeneration of photoreceptors caused by excessive illumination, inherited mutations, or aging is the principal pathology of blinding diseases. Pharmacological compounds that stabilize the visual receptor rhodopsin and modulate the cellular pathways triggering death of photoreceptors could avert this pathology. Interestingly, flavonoids can modulate the cellular processes, such as oxidative stress, inflammatory responses, and apoptosis, that are activated during retinal degeneration. As we found previously, flavonoids also bind directly to unliganded rod opsin, enhancing its folding, stability, and regeneration. In addition, flavonoids stimulate rhodopsin gene expression. Thus, we evaluated the effect of two main dietary flavonoids, quercetin and myricetin, in ATP-binding cassette subfamily A member 4 -/- /retinol dehydrogenase 8 -/- and wild-type BALB/c mice susceptible to light-induced photoreceptor degeneration. Using in vivo imaging, such as optical coherence tomography, scanning laser ophthalmoscopy, and histologic assessment of retinal morphology, we found that treatment with these flavonoids prior to light insult remarkably protected retina from deterioration and preserved its function. Using high-performance liquid chromatography-mass spectrometry analysis, we detected these flavonoids in the eye upon their intraperitoneal administration. The molecular events associated with the protective effect of quercetin and myricetin were related to the elevated expression of photoreceptor-specific proteins, rhodopsin and cone opsins, decreased expression of the specific inflammatory markers, and the shift of the equilibrium between cell death regulators BCL2-associated X protein (BAX) and B-cell lymphoma 2 toward an antiapoptotic profile. These results were confirmed in photoreceptor-derived 661W cells treated with either H2O2 or all-trans-retinal stressors implicated in the mechanism of retinal degeneration. Altogether, flavonoids could have significant prophylactic value for retinal degenerative diseases. SIGNIFICANCE STATEMENT: Flavonoids commonly present in food exhibit advantageous effects in blinding diseases. They bind to and stabilize unliganded rod opsin, which in excess accelerates degenerative processes in the retina. Additionally, flavonoids enhance the expression of the visual receptors, rod and cone opsins; inhibit the inflammatory reactions; and induce the expression of antiapoptotic markers in the retina, preventing the degeneration in vivo. Thus, flavonoids could have a prophylactic value for retinal degenerative diseases.
Collapse
Affiliation(s)
- Joseph T Ortega
- Department of Pharmacology, Cleveland Center for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Tanu Parmar
- Department of Pharmacology, Cleveland Center for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Marcin Golczak
- Department of Pharmacology, Cleveland Center for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Beata Jastrzebska
- Department of Pharmacology, Cleveland Center for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
53
|
Bucher K, Rodríguez-Bocanegra E, Dauletbekov D, Fischer MD. Immune responses to retinal gene therapy using adeno-associated viral vectors - Implications for treatment success and safety. Prog Retin Eye Res 2020; 83:100915. [PMID: 33069860 DOI: 10.1016/j.preteyeres.2020.100915] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 10/08/2020] [Accepted: 10/10/2020] [Indexed: 02/06/2023]
Abstract
Recombinant adeno-associated virus (AAV) is the leading vector for gene therapy in the retina. As non-pathogenic, non-integrating, replication deficient vector, the recombinant virus efficiently transduces all key retinal cell populations. Successful testing of AAV vectors in clinical trials of inherited retinal diseases led to the recent approval of voretigene neparvovec (Luxturna) for the treatment of RPE65 mutation-associated retinal dystrophies. However, studies applying AAV-mediated retinal gene therapy independently reported intraocular inflammation and/or loss of efficacy after initial functional improvements. Both observations might be explained by targeted removal of transduced cells via anti-viral defence mechanisms. AAV has been shown to activate innate pattern recognition receptors (PRRs) such as toll-like receptor (TLR)-2 and TLR-9 resulting in the release of inflammatory cytokines and type I interferons. The vector can also induce capsid-specific and transgene-specific T cell responses and neutralizing anti-AAV antibodies which both limit the therapeutic effect. However, the target organ of retinal gene therapy, the eye, is known as an immune-privileged site. It is characterized by suppression of inflammation and promotion of immune tolerance which might prevent AAV-induced immune responses. This review evaluates AAV-related immune responses, toxicity and inflammation in studies of retinal gene therapy, identifies influencing variables of these responses and discusses potential strategies to modulate immune reactions to AAV vectors to increase the safety and efficacy of ocular gene therapy.
Collapse
Affiliation(s)
- Kirsten Bucher
- University Eye Hospital, Centre for Ophthalmology, University Hospital Tübingen, Tübingen, Germany; Institute for Ophthalmic Research, Centre for Ophthalmology, University Hospital Tübingen, Tübingen, Germany
| | - Eduardo Rodríguez-Bocanegra
- University Eye Hospital, Centre for Ophthalmology, University Hospital Tübingen, Tübingen, Germany; Institute for Ophthalmic Research, Centre for Ophthalmology, University Hospital Tübingen, Tübingen, Germany
| | - Daniyar Dauletbekov
- University Eye Hospital, Centre for Ophthalmology, University Hospital Tübingen, Tübingen, Germany; Institute for Ophthalmic Research, Centre for Ophthalmology, University Hospital Tübingen, Tübingen, Germany
| | - M Dominik Fischer
- University Eye Hospital, Centre for Ophthalmology, University Hospital Tübingen, Tübingen, Germany; Institute for Ophthalmic Research, Centre for Ophthalmology, University Hospital Tübingen, Tübingen, Germany; Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK; Nuffield Laboratory of Ophthalmology, Department of Clinical Neurosciences, University of Oxford, Oxford, UK.
| |
Collapse
|
54
|
Cheng X, He D, Liao C, Lin S, Tang L, Wang YL, Hu J, Li W, Liu Z, Wu Y, Liao Y. IL-1/IL-1R signaling induced by all-trans-retinal contributes to complement alternative pathway activation in retinal pigment epithelium. J Cell Physiol 2020; 236:3660-3674. [PMID: 33034385 DOI: 10.1002/jcp.30103] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 09/27/2020] [Accepted: 09/29/2020] [Indexed: 12/23/2022]
Abstract
The underlying mechanisms of complement activation in Stargardt disease type 1 (STGD1) and age-related macular degeneration (AMD) are not fully understood. Overaccumulation of all-trans-retinal (atRAL) has been proposed as the pathogenic factor in both diseases. By incubating retinal pigment epithelium (RPE) cells with atRAL, we showed that C5b-9 membrane attack complexes (MACs) were generated mainly through complement alternative pathway. An increase in complement factor B (CFB) expression as well as downregulation of complement regulatory proteins CD46, CD55, CD59, and CFH were observed in RPE cells after atRAL treatment. Furthermore, interleukin-1β production was provoked in both atRAL-treated RPE cells and microglia/macrophages. Coincubation of RPE cells with interleukin-1 receptor antagonist (IL1Ra) and atRAL ameliorated complement activation and downregulated CFB expression by attenuating both p38 and c-Jun N-terminal kinase (JNK) signaling pathways. Our findings demonstrate that atRAL induces an autocrine/paracrine IL-1/IL-1R signaling to promote complement alternative pathway activation in RPE cells and provide a novel perspective on the pathomechanism of macular degeneration.
Collapse
Affiliation(s)
- Xinxuan Cheng
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Eye Institute of Xiamen University, Xiamen University, Xiamen, China.,Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, China.,Department of Ophthalmology, Xiamen University Affiliated Xiamen Eye Center, Xiamen, China
| | - Danxue He
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Eye Institute of Xiamen University, Xiamen University, Xiamen, China.,Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, China.,Department of Ophthalmology, Xiamen University Affiliated Xiamen Eye Center, Xiamen, China
| | - Chunyan Liao
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Eye Institute of Xiamen University, Xiamen University, Xiamen, China.,Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, China.,Department of Ophthalmology, Xiamen University Affiliated Xiamen Eye Center, Xiamen, China
| | - Sijie Lin
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Eye Institute of Xiamen University, Xiamen University, Xiamen, China.,Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, China.,Department of Ophthalmology, Xiamen University Affiliated Xiamen Eye Center, Xiamen, China
| | - Liying Tang
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Eye Institute of Xiamen University, Xiamen University, Xiamen, China.,Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, China.,Department of Ophthalmology, Xiamen University Affiliated Xiamen Eye Center, Xiamen, China
| | - Yuan-Liang Wang
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, China.,Section of Molecular Biology, University of California, San Diego, La Jolla, California, USA
| | - Jiaoyue Hu
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Eye Institute of Xiamen University, Xiamen University, Xiamen, China.,Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, China.,Department of Ophthalmology, Xiamen University Affiliated Xiamen Eye Center, Xiamen, China
| | - Wei Li
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Eye Institute of Xiamen University, Xiamen University, Xiamen, China.,Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, China.,Department of Ophthalmology, Xiamen University Affiliated Xiamen Eye Center, Xiamen, China
| | - Zuguo Liu
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Eye Institute of Xiamen University, Xiamen University, Xiamen, China.,Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, China.,Department of Ophthalmology, Xiamen University Affiliated Xiamen Eye Center, Xiamen, China
| | - Yalin Wu
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Eye Institute of Xiamen University, Xiamen University, Xiamen, China.,Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, China.,Department of Ophthalmology, Xiamen University Affiliated Xiamen Eye Center, Xiamen, China
| | - Yi Liao
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Eye Institute of Xiamen University, Xiamen University, Xiamen, China.,Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, China.,Department of Ophthalmology, Xiamen University Affiliated Xiamen Eye Center, Xiamen, China
| |
Collapse
|
55
|
Federico S, Pozzetti L, Papa A, Carullo G, Gemma S, Butini S, Campiani G, Relitti N. Modulation of the Innate Immune Response by Targeting Toll-like Receptors: A Perspective on Their Agonists and Antagonists. J Med Chem 2020; 63:13466-13513. [PMID: 32845153 DOI: 10.1021/acs.jmedchem.0c01049] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Toll-like receptors (TLRs) are a class of proteins that recognize pathogen-associated molecular patterns (PAMPs) and damaged-associated molecular patterns (DAMPs), and they are involved in the regulation of innate immune system. These transmembrane receptors, localized at the cellular or endosomal membrane, trigger inflammatory processes through either myeloid differentiation primary response 88 (MyD88) or TIR-domain-containing adapter-inducing interferon-β (TRIF) signaling pathways. In the last decades, extensive research has been performed on TLR modulators and their therapeutic implication under several pathological conditions, spanning from infections to cancer, from metabolic disorders to neurodegeneration and autoimmune diseases. This Perspective will highlight the recent discoveries in this field, emphasizing the role of TLRs in different diseases and the therapeutic effect of their natural and synthetic modulators, and it will discuss insights for the future exploitation of TLR modulators in human health.
Collapse
Affiliation(s)
- Stefano Federico
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022, University of Siena, via Aldo Moro 2, 53100, Siena, Italy
| | - Luca Pozzetti
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022, University of Siena, via Aldo Moro 2, 53100, Siena, Italy
| | - Alessandro Papa
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022, University of Siena, via Aldo Moro 2, 53100, Siena, Italy
| | - Gabriele Carullo
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022, University of Siena, via Aldo Moro 2, 53100, Siena, Italy
| | - Sandra Gemma
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022, University of Siena, via Aldo Moro 2, 53100, Siena, Italy
| | - Stefania Butini
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022, University of Siena, via Aldo Moro 2, 53100, Siena, Italy
| | - Giuseppe Campiani
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022, University of Siena, via Aldo Moro 2, 53100, Siena, Italy
| | - Nicola Relitti
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022, University of Siena, via Aldo Moro 2, 53100, Siena, Italy
| |
Collapse
|
56
|
Karlen SJ, Miller EB, Burns ME. Microglia Activation and Inflammation During the Death of Mammalian Photoreceptors. Annu Rev Vis Sci 2020; 6:149-169. [PMID: 32936734 PMCID: PMC10135402 DOI: 10.1146/annurev-vision-121219-081730] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Photoreceptors are highly specialized sensory neurons with unique metabolic and physiological requirements. These requirements are partially met by Müller glia and cells of the retinal pigment epithelium (RPE), which provide essential metabolites, phagocytose waste, and control the composition of the surrounding microenvironment. A third vital supporting cell type, the retinal microglia, can provide photoreceptors with neurotrophic support or exacerbate neuroinflammation and hasten neuronal cell death. Understanding the physiological requirements for photoreceptor homeostasis and the factors that drive microglia to best promote photoreceptor survival has important implications for the treatment and prevention of blinding degenerative diseases like retinitis pigmentosa and age-related macular degeneration.
Collapse
Affiliation(s)
- Sarah J. Karlen
- Department of Cell Biology and Human Anatomy, University of California, Davis, Davis, California 95616, USA
| | - Eric B. Miller
- Center for Neuroscience, University of California, Davis, Davis, California 95616, USA
| | - Marie E. Burns
- Department of Cell Biology and Human Anatomy, University of California, Davis, Davis, California 95616, USA
- Center for Neuroscience, University of California, Davis, Davis, California 95616, USA
- Department of Ophthalmology & Vision Science, University of California, Davis, Davis, California 95616, USA
| |
Collapse
|
57
|
Makabe K, Sugita S, Mandai M, Futatsugi Y, Takahashi M. Microglia dynamics in retinitis pigmentosa model: formation of fundus whitening and autofluorescence as an indicator of activity of retinal degeneration. Sci Rep 2020; 10:14700. [PMID: 32895435 PMCID: PMC7477572 DOI: 10.1038/s41598-020-71626-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Accepted: 06/04/2020] [Indexed: 01/04/2023] Open
Abstract
In patients with retinitis pigmentosa (RP), color fundus photography and fundus autofluorescence (FAF) have been used to estimate the disease progression. To understand the origin and the diagnostic interpretation of the fundus color and FAF, we performed in vivo imaging of fundus color and FAF together with histological analyses of the retinal degeneration process using the RP model mice, rd10. FAF partly represented the accumulation of microglia in the photoreceptor outer segments. Fundus whitening suggested the presence of apoptotic cells, which spatiotemporally preceded increase in FAF. We observed two patterns of FAF localization, arcuate and diffuse, each indicating different pattern of apoptosis, wavy and diffuse, respectively. Diffuse pattern of apoptosis was suppressed in dark-raised rd10 mice, in which outer nuclear layer (ONL) loss was significantly suppressed. The occupancy of FAF correlated with the thinning rate of the ONL. Fractalkine, a microglia chemotactic factor, was detected in apoptotic photoreceptors, suggesting chemokine-induced recruitment of microglia into the ONL, which paralleled with accelerated ONL loss and increased FAF occupancy. Thus, we propose that the degree of photoreceptor apoptosis and the rate of ONL thinning in RP patients might be read from the fundus color and the FAF.
Collapse
Affiliation(s)
- Kenichi Makabe
- Laboratory for Retinal Regeneration, Center for Biosystems Dynamics Research, RIKEN, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan.,Department of Ophthalmology, Kobe Kaisei Hospital, 3-11-15 Shinoharakitamachi, Nada-ku, Kobe, 657-0068, Japan
| | - Sunao Sugita
- Laboratory for Retinal Regeneration, Center for Biosystems Dynamics Research, RIKEN, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan.
| | - Michiko Mandai
- Laboratory for Retinal Regeneration, Center for Biosystems Dynamics Research, RIKEN, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan
| | - Yoko Futatsugi
- Laboratory for Retinal Regeneration, Center for Biosystems Dynamics Research, RIKEN, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan
| | - Masayo Takahashi
- Laboratory for Retinal Regeneration, Center for Biosystems Dynamics Research, RIKEN, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan
| |
Collapse
|
58
|
Arslan U. Management of cystoid macular edema secondary to retinitis pigmentosa via subliminal micropulse yellow laser. Lasers Med Sci 2020; 36:317-323. [PMID: 32363437 DOI: 10.1007/s10103-020-03031-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Accepted: 04/22/2020] [Indexed: 11/30/2022]
Abstract
To investigate the effects of subliminal micropulse yellow laser application on the central macular thickness and best-corrected visual acuity in cystoid macular edema secondary to retinitis pigmentosa patients. This prospective open-label clinical trial, conducted between January 2018 and October 2019, included 32 eyes of 29 patients who had cystoid macular edema secondary to retinitis pigmentosa. Patients were treated by subliminal micropulse yellow laser for one session. Central macular thickness and best-corrected visual acuity changes were investigated just before the treatment and 1 year later after the one session of the treatment. The mean central macular thickness was 651.3 μm before the treatment and 247.7 μm at 12 months after the treatment. The decrease in mean central macular thickness was statistically significant (p = 0.01). Median best-corrected visual acuity was 66.8 ETDRS letters before the treatment and 70.0 letters at 12 months after the treatment. The increase in best-corrected visual acuity was not statistically significant (p = 0.18). Eighty-six percent of the patients stated that the quality of central vision increased and that color vision, contrast sensitivity, and distortion improved. We did not encounter any serious adverse events related to the application of subliminal micropulse yellow laser. The subliminal micropulse yellow laser seems to be a therapeutic, effective, and safe option for the treatment of non-inflammatory and resistant cystoid macular edema secondary to retinitis pigmentosa patients. ClinicalTrials.gov ID: NCT04234438, January 17, 2020.
Collapse
Affiliation(s)
- Umut Arslan
- Ankara University Technopolis, Bioretina, Ankara, Turkey. .,Neorama Ofis 55-56 Yaşam Cad, No 13/A Beştepe /Yenimahalle, Ankara, Turkey.
| |
Collapse
|
59
|
Fang Y, Tschulakow A, Taubitz T, Illing B, Biesemeier A, Julien-Schraermeyer S, Radu RA, Jiang Z, Schraermeyer U. Fundus autofluorescence, spectral-domain optical coherence tomography, and histology correlations in a Stargardt disease mouse model. FASEB J 2020; 34:3693-3714. [PMID: 31989709 DOI: 10.1096/fj.201901784rr] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 12/17/2019] [Accepted: 12/17/2019] [Indexed: 01/09/2023]
Abstract
Stargardt disease (STGD1), known as inherited retinal dystrophy, is caused by ABCA4 mutations. The pigmented Abca4-/- mouse strain only reflects the early stage of STGD1 since it is devoid of retinal degeneration. This blue light-illuminated pigmented Abca4-/- mouse model presented retinal pigment epithelium (RPE) and photoreceptor degeneration which was similar to the advanced STGD1 phenotype. In contrast, wild-type mice showed no RPE degeneration after blue light illumination. In Abca4-/- mice, the acute blue light diminished the mean autofluorescence (AF) intensity in both fundus short-wavelength autofluorescence (SW-AF) and near-infrared autofluorescence (NIR-AF) modalities correlating with reduced levels of bisretinoid-fluorophores. Blue light-induced RPE cellular damage preceded the photoreceptors loss. In late-stage STGD1-like patient and blue light-illuminated Abca4-/- mice, lipofuscin and melanolipofuscin granules were found to contribute to NIR-AF, indicated by the colocalization of lipofuscin-AF and NIR-AF under the fluorescence microscope. In this mouse model, the correlation between in vivo and ex vivo assessments revealed histological characteristics of fundus AF abnormalities. The flecks which are hyper AF in both SW-AF and NIR-AF corresponded to the subretinal macrophages fully packed with pigment granules (lipofuscin, melanin, and melanolipofuscin). This mouse model, which has the phenotype of advanced STGD1, is important to understand the histopathology of Stargardt disease.
Collapse
Affiliation(s)
- Yuan Fang
- Division of Experimental Vitreoretinal Surgery, Center for Ophthalmology, Institute of Ophthalmic Research, University of Tuebingen, Tuebingen, Germany
| | - Alexander Tschulakow
- Division of Experimental Vitreoretinal Surgery, Center for Ophthalmology, Institute of Ophthalmic Research, University of Tuebingen, Tuebingen, Germany
- Preclinical Drug Assessment, STZ Ocutox, Hechingen, Germany
| | - Tatjana Taubitz
- Division of Experimental Vitreoretinal Surgery, Center for Ophthalmology, Institute of Ophthalmic Research, University of Tuebingen, Tuebingen, Germany
| | - Barbara Illing
- Division of Experimental Vitreoretinal Surgery, Center for Ophthalmology, Institute of Ophthalmic Research, University of Tuebingen, Tuebingen, Germany
| | - Antje Biesemeier
- Division of Experimental Vitreoretinal Surgery, Center for Ophthalmology, Institute of Ophthalmic Research, University of Tuebingen, Tuebingen, Germany
| | - Sylvie Julien-Schraermeyer
- Division of Experimental Vitreoretinal Surgery, Center for Ophthalmology, Institute of Ophthalmic Research, University of Tuebingen, Tuebingen, Germany
- Preclinical Drug Assessment, STZ Ocutox, Hechingen, Germany
| | - Roxana A Radu
- UCLA Stein Eye Institute, Department of Ophthalmology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Zhichun Jiang
- UCLA Stein Eye Institute, Department of Ophthalmology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Ulrich Schraermeyer
- Division of Experimental Vitreoretinal Surgery, Center for Ophthalmology, Institute of Ophthalmic Research, University of Tuebingen, Tuebingen, Germany
- Preclinical Drug Assessment, STZ Ocutox, Hechingen, Germany
| |
Collapse
|
60
|
Increased Neuroprotective Microglia and Photoreceptor Survival in the Retina from a Peptide Inhibitor of Myeloid Differentiation Factor 88 (MyD88). J Mol Neurosci 2020; 70:968-980. [PMID: 32072483 DOI: 10.1007/s12031-020-01503-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 02/05/2020] [Indexed: 01/14/2023]
Abstract
Myeloid differentiation factor 88 (MyD88) is an adaptor protein for the Toll-like receptor (TLR) and interleukin 1 receptor (IL-1R) families of innate immunity receptors that mediate inflammatory responses to cellular injury. TLR/IL1R/MyD88 signaling is known to contribute to retinal degeneration, although how MyD88 regulates neuronal survival, and the effect of MyD88 on the inflammatory environment in the retina, is mostly unknown. In this study, we tested the hypothesis that blocking MyD88-mediated signaling early in retinal degeneration promotes transition of microglia towards a neuroprotective anti-inflammatory phenotype, resulting in enhanced photoreceptor survival. We also tested whether systemic delivery of a pharmacologic MyD88 inhibitor has therapeutic potential. The rd10 mouse model of retinal degeneration was injected intraperitoneally with increasing doses of a MyD88 blocking peptide or control peptide early in degeneration, and inflammatory responses and photoreceptor survival were measured at specific time points using flow cytometry, cytokine profiling, and electroretinograms. Our results demonstrated that rd10 mice injected with a low dose of MyD88 inhibitor peptide showed increased rod photoreceptor function and reduced apoptosis compared with control peptide and uninjected mice. MyD88 inhibition also resulted in fewer microglia/macrophage cells in the photoreceptor layer whereas total peripheral and retinal macrophage were not changed. Furthermore, increased number of cells expressing the Arg1 marker of neuroprotective microglia in the photoreceptor layer and higher MCP-1 and anti-inflammatory cytokine IL-27 were associated with photoreceptor survival. Therefore, these data suggest that the MyD88 inhibitor modified the retina environment to become less inflammatory, leading to improved photoreceptor function and survival.
Collapse
|
61
|
Inhibition of LOX-1 prevents inflammation and photoreceptor cell death in retinal degeneration. Int Immunopharmacol 2020; 80:106190. [PMID: 31945611 DOI: 10.1016/j.intimp.2020.106190] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 12/10/2019] [Accepted: 01/03/2020] [Indexed: 11/21/2022]
Abstract
PURPOSE To explore the expression and role of lectin-like oxidized low-density lipoprotein receptor 1 (LOX-1) in retinal degeneration. METHODS The retinal degeneration of BALB/c mice was induced by light exposure. BV2 cells were activated by LPS stimulation. Retinas or BV2 cells were pretreated with LOX-1 neutralizing antibody or Polyinosinic acid (PolyI) (the inhibitor of LOX-1) before light damage (LD) or LPS stimulation. LOX-1, TNF-α, IL-1β, CCL2 and NF-κB expression were detected in retinas or BV2 cells by real-time RT-PCR, western blot or ELISA. Histological analyses of retinas were performed. Photoreceptor cell death was assessed by TUNEL assay in retinas or by flow cytometry in 661W cells cultured in microglia-conditioned medium. RESULTS Photoreceptor cell death and elevated expression of LOX-1 were induced by LD in retinas of BALB/c mice. LOX-1 neutralizing antibody or PolyI pretreatment significantly reduced the elevated expression of LOX-1, TNF-α, IL-1β, CCL2 and p-NF-κB caused by LD in retinas. Inhibition of LOX-1 by LOX-1 neutralizing antibody or PolyI significantly reduced photoreceptor cell death induced by LD in retinas. Elevated levels of TNF-α, IL-1β and CCL2 caused by LPS were down-regulated by inhibition of LOX-1 in BV2 cells. Inhibition of LOX-1 reduces microglial neurotoxicity on photoreceptors. CONCLUSIONS LOX-1 expression is increased in light induced retinal degeneration, what's more, inhibition of LOX-1 prevents inflammation and photoreceptor cell death in retinal degeneration and reduces microglial neurotoxicity on photoreceptors. Therefore, LOX-1 can be used as a potential therapeutic target for such retinal degeneration diseases.
Collapse
|
62
|
Lavalette S, Conart JB, Touhami S, Roubeix C, Houssier M, Augustin S, Raoul W, Combadière C, Febbraio M, Ong H, Chemtob S, Sahel JA, Delarasse C, Guillonneau X, Sennlaub F. CD36 Deficiency Inhibits Retinal Inflammation and Retinal Degeneration in Cx3cr1 Knockout Mice. Front Immunol 2020; 10:3032. [PMID: 31969887 PMCID: PMC6960398 DOI: 10.3389/fimmu.2019.03032] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 12/10/2019] [Indexed: 12/22/2022] Open
Abstract
Background: CD36, a member of the class B scavenger receptor family, participates in Toll-like receptor signaling on mononuclear phagocytes (MP) and can promote sterile pathogenic inflammation. We here analyzed the effect of CD36 deficiency on retinal inflammation and photoreceptor degeneration, the hallmarks of age-related macular degeneration (AMD), that characterize Cx3cr1−/−mice. Methods: We analyzed subretinal MP accumulation, and cone- and rod-degeneration in light-challenged and aged, CD36 competent or deficient, hyper-inflammatory Cx3cr1−/− mice, using histology and immune-stained retinal flatmounts. Monocytes (Mo) were subretinally adoptively transferred to evaluate their elimination rate from the subretinal space and Interleukin 6 (IL-6) secretion from cultured Mo-derived cells (MdCs) of the different mouse strains were analyzed. Results: CD36 deficient Cx3cr1−/− mice were protected against age- and light-induced subretinal inflammation and associated cone and rod degeneration. CD36 deficiency in Cx3cr1−/− MPs inhibited their prolonged survival in the immune-suppressive subretinal space and reduced the exaggerated IL-6 secretion observed in Cx3cr1−/− MPs that we previously showed leads to increased subretinal MP survival. Conclusion:Cd36 deficiency significantly protected hyperinflammatory Cx3cr1−/− mice against subretinal MP accumulation and associated photoreceptor degeneration. The observed CD36-dependent induction of pro-inflammatory IL-6 might be at least partially responsible for the prolonged MP survival in the immune-suppressive environment and its pathological consequences on photoreceptor homeostasis.
Collapse
Affiliation(s)
- Sophie Lavalette
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France
| | | | - Sara Touhami
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France
| | | | - Marianne Houssier
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France
| | | | - William Raoul
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France.,Université de Tours, Inserm, N2C UMR 1069, Faculté de Médecine, Tours, France
| | - Christophe Combadière
- Sorbonne Université, Inserm, CNRS, Centre d'Immunologie et des Maladies Infectieuses, Cimi-Paris, Paris, France
| | - Maria Febbraio
- Department of Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Huy Ong
- Faculty of Pharmacy, Université de Montréal, Montreal, QC, Canada
| | - Sylvain Chemtob
- Departments of Pediatrics, Ophthalmology and Pharmacology, Université de Montréal, Montreal, QC, Canada
| | - José-Alain Sahel
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France
| | - Cécile Delarasse
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France
| | | | - Florian Sennlaub
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France
| |
Collapse
|
63
|
Charles-Messance H, Blot G, Couturier A, Vignaud L, Touhami S, Beguier F, Siqueiros L, Forster V, Barmo N, Augustin S, Picaud S, Sahel JA, Rendon A, Grosche A, Tadayoni R, Sennlaub F, Guillonneau X. IL-1β induces rod degeneration through the disruption of retinal glutamate homeostasis. J Neuroinflammation 2020; 17:1. [PMID: 31900165 PMCID: PMC6942287 DOI: 10.1186/s12974-019-1655-5] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 11/21/2019] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Age-related macular degeneration is characterized by the accumulation of subretinal macrophages and the degeneration of cones, but mainly of rods. We have previously shown that Mononuclear Phagocytes-derived IL-1β induces rod photoreceptor cell death during experimental subretinal inflammation and in retinal explants exposed to IL-1β but the mechanism is unknown. METHODS Retinal explants were culture in the presence of human monocytes or IL-1β and photoreceptor cell survival was analyzed by TUNEL labeling. Glutamate concentration and transcription levels of gene involved in the homeostasis of glutamate were analyzed in cell fractions of explant cultured or not in the presence of IL-1β. Glutamate receptor antagonists were evaluated for their ability to reduce photoreceptor cell death in the presence of IL1-β or monocytes. RESULTS We here show that IL-1β does not induce death in isolated photoreceptors, suggesting an indirect effect. We demonstrate that IL-1β leads to glutamate-induced rod photoreceptor cell death as it increases the extracellular glutamate concentrations in the retina through the inhibition of its conversion to glutamine in Müller cells, increased release from Müller cells, and diminished reuptake. The inhibition of non-NMDA receptors completely and efficiently prevented rod apoptosis in retinal explants cultured in the presence of IL-1β or, more importantly, in vivo, in a model of subretinal inflammation. CONCLUSIONS Our study emphasizes the importance of inflammation in the deregulation of glutamate homeostasis and provides a comprehensive mechanism of action for IL-1β-induced rod degeneration.
Collapse
Affiliation(s)
- Hugo Charles-Messance
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012, Paris, France
| | - Guillaume Blot
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012, Paris, France
| | - Aude Couturier
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012, Paris, France.,Department of Ophthalmology, Hôpital Lariboisière, Paris, France
| | - Lucile Vignaud
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012, Paris, France
| | - Sara Touhami
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012, Paris, France.,Department of Ophthalmology, Hôpital Lariboisière, Paris, France
| | - Fanny Beguier
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012, Paris, France
| | - Lourdes Siqueiros
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012, Paris, France
| | - Valérie Forster
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012, Paris, France
| | - Nour Barmo
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012, Paris, France
| | - Sébastien Augustin
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012, Paris, France
| | - Serge Picaud
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012, Paris, France
| | - José-Alain Sahel
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012, Paris, France.,CHNO des Quinze-Vingts, DHU Sight Restore, INSERM-DGOS CIC 1423, 28 rue de Charenton, F-75012, Paris, France.,Department of Ophthalmology, The University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Alvaro Rendon
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012, Paris, France
| | - Antje Grosche
- Department of Physiological Genomics, Ludwig-Maximilians-Universität München, Grosshaderner Str. 9, D-82152, Planegg-Martinsried, Germany
| | - Ramin Tadayoni
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012, Paris, France.,Department of Ophthalmology, Hôpital Lariboisière, Paris, France
| | - Florian Sennlaub
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012, Paris, France
| | - Xavier Guillonneau
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012, Paris, France.
| |
Collapse
|
64
|
Will-Orrego A, Qiu Y, Fassbender ES, Shen S, Aranda J, Kotagiri N, Maker M, Liao SM, Jaffee BD, Poor SH. Amount of Mononuclear Phagocyte Infiltrate Does Not Predict Area of Experimental Choroidal Neovascularization (CNV). J Ocul Pharmacol Ther 2019; 34:489-499. [PMID: 30188257 PMCID: PMC6152860 DOI: 10.1089/jop.2017.0131] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Purpose: Mononuclear phagocytes (MNPs) are present in neovascular age-related macular degeneration (nv AMD) which is also called choroidal neovascularization (CNV). The number and phenotype of the MNPs depend upon the local environment in the CNV and effect of nv AMD therapy. We investigated ocular cell infiltration and conditions that modulate angiogenesis in a laser-induced mouse CNV model. Methods: We developed assays to quantify MNPs in our established mouse CNV model. One MNP assay quantified the number of subretinal cells peripheral to the CNV lesions. A second assay semiquantitatively assesses the number of MNPs localized to the CNV lesion. We used these assays to measure the effect of toll-like receptor-2 (TLR-2) activation, anti-vascular endothelial growth factor (VEGF) therapy, and chemokine (C-C motif) ligand 2 (Ccl2) genetic deletion on MNP infiltration after laser injury. Results: Laser injury induced blood vessel growth and infiltration of MNPs. Systemic administration of a TLR-2 activating peptide increased laser-induced CNV area, MNP cell numbers, and MNP density over the CNV lesions. Systemic administration of a VEGF antibody reduced CNV area, while Ccl2 genetic deletion increased CNV area. Despite the change in amount of angiogenesis, MNP infiltration was, surprisingly, unchanged in these 2 conditions. Conclusions: MNP quantification provides biological insights for candidate AMD therapies. The number of infiltrating MNP cells does not correlate with the amount of laser-induced CNV area.
Collapse
Affiliation(s)
- Adrian Will-Orrego
- Department of Ophthalmology, Novartis Institutes for Biomedical Research , Cambridge, Massachusetts
| | - Yubin Qiu
- Department of Ophthalmology, Novartis Institutes for Biomedical Research , Cambridge, Massachusetts
| | - Elizabeth S Fassbender
- Department of Ophthalmology, Novartis Institutes for Biomedical Research , Cambridge, Massachusetts
| | - Siyuan Shen
- Department of Ophthalmology, Novartis Institutes for Biomedical Research , Cambridge, Massachusetts
| | - Jorge Aranda
- Department of Ophthalmology, Novartis Institutes for Biomedical Research , Cambridge, Massachusetts
| | - Namrata Kotagiri
- Department of Ophthalmology, Novartis Institutes for Biomedical Research , Cambridge, Massachusetts
| | - Michael Maker
- Department of Ophthalmology, Novartis Institutes for Biomedical Research , Cambridge, Massachusetts
| | - Sha-Mei Liao
- Department of Ophthalmology, Novartis Institutes for Biomedical Research , Cambridge, Massachusetts
| | - Bruce D Jaffee
- Department of Ophthalmology, Novartis Institutes for Biomedical Research , Cambridge, Massachusetts
| | - Stephen H Poor
- Department of Ophthalmology, Novartis Institutes for Biomedical Research , Cambridge, Massachusetts
| |
Collapse
|
65
|
Wooff Y, Man SM, Aggio-Bruce R, Natoli R, Fernando N. IL-1 Family Members Mediate Cell Death, Inflammation and Angiogenesis in Retinal Degenerative Diseases. Front Immunol 2019; 10:1618. [PMID: 31379825 PMCID: PMC6646526 DOI: 10.3389/fimmu.2019.01618] [Citation(s) in RCA: 152] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 06/28/2019] [Indexed: 12/22/2022] Open
Abstract
Inflammation underpins and contributes to the pathogenesis of many retinal degenerative diseases. The recruitment and activation of both resident microglia and recruited macrophages, as well as the production of cytokines, are key contributing factors for progressive cell death in these diseases. In particular, the interleukin 1 (IL-1) family consisting of both pro- and anti-inflammatory cytokines has been shown to be pivotal in the mediation of innate immunity and contribute directly to a number of retinal degenerations, including Age-Related Macular Degeneration (AMD), diabetic retinopathy, retinitis pigmentosa, glaucoma, and retinopathy of prematurity (ROP). In this review, we will discuss the role of IL-1 family members and inflammasome signaling in retinal degenerative diseases, piecing together their contribution to retinal disease pathology, and identifying areas of research expansion required to further elucidate their function in the retina.
Collapse
Affiliation(s)
- Yvette Wooff
- The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia.,ANU Medical School, The Australian National University, Canberra, ACT, Australia
| | - Si Ming Man
- The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Riemke Aggio-Bruce
- The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Riccardo Natoli
- The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia.,ANU Medical School, The Australian National University, Canberra, ACT, Australia
| | - Nilisha Fernando
- The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| |
Collapse
|
66
|
Getter T, Suh S, Hoang T, Handa JT, Dong Z, Ma X, Chen Y, Blackshaw S, Palczewski K. The selective estrogen receptor modulator raloxifene mitigates the effect of all- trans-retinal toxicity in photoreceptor degeneration. J Biol Chem 2019; 294:9461-9475. [PMID: 31073029 DOI: 10.1074/jbc.ra119.008697] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 05/08/2019] [Indexed: 12/19/2022] Open
Abstract
The retinoid cycle is a metabolic process in the vertebrate retina that continuously regenerates 11-cis-retinal (11-cisRAL) from the all-trans-retinal (atRAL) isomer. atRAL accumulation can cause photoreceptor degeneration and irreversible visual dysfunction associated with incurable blinding retinal diseases, such as Stargardt disease, retinitis pigmentosa (RP), and atrophic age-related macular degeneration (AMD). The underlying cellular mechanisms leading to retinal degeneration remain uncertain, although previous studies have shown that atRAL promotes calcium influx associated with cell apoptosis. To identify compounds that mitigate the effects of atRAL toxicity, here we developed an unbiased and robust image-based assay that can detect changes in intracellular calcium levels in U2OS cells. Using our assay in a high-throughput screen of 2,400 compounds, we noted that selective estrogen receptor modulators (SERMs) potently stabilize intracellular calcium and thereby counteract atRAL-induced toxicity. In a light-induced retinal degeneration mouse model (Abca4 -/- Rdh8 -/-), raloxifene (a benzothiophene-type scaffold SERM) prevented the onset of photoreceptor apoptosis and thus protected the retina from degeneration. The minor structural differences between raloxifene and one of its derivatives (Y 134) had a major impact on calcium homeostasis after atRAL exposure in vitro, and we verified this differential impact in vivo In summary, the SERM raloxifene has structural and functional neuroprotective effects in the retina. We propose that the highly sensitive image-based assay developed here could be applied for the discovery of additional drug candidates preventing photoreceptor degeneration.
Collapse
Affiliation(s)
- Tamar Getter
- From the Gavin Herbert Eye Institute, Department of Ophthalmology, University of California, Irvine, California 92697, .,the Department of Pharmacology, Case Western Reserve University, Cleveland, Ohio 44106
| | - Susie Suh
- From the Gavin Herbert Eye Institute, Department of Ophthalmology, University of California, Irvine, California 92697.,the Department of Pharmacology, Case Western Reserve University, Cleveland, Ohio 44106
| | - Thanh Hoang
- the Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - James T Handa
- the Wilmer Eye Institute, Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287
| | | | - Xiuli Ma
- Polgenix Inc., Irvine, California 92617
| | - Yuanyuan Chen
- the Department of Ophthalmology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, and.,the McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15213
| | - Seth Blackshaw
- the Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Krzysztof Palczewski
- From the Gavin Herbert Eye Institute, Department of Ophthalmology, University of California, Irvine, California 92697, .,the Department of Pharmacology, Case Western Reserve University, Cleveland, Ohio 44106
| |
Collapse
|
67
|
Massengill MT, Ahmed CM, Lewin AS, Ildefonso CJ. Neuroinflammation in Retinitis Pigmentosa, Diabetic Retinopathy, and Age-Related Macular Degeneration: A Minireview. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1074:185-191. [PMID: 29721943 DOI: 10.1007/978-3-319-75402-4_23] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The eye is an immuno-privileged organ. However, certain diseases such as uveitis are intrinsically linked to inflammation. In several retinal degenerative diseases, there is a unique damage at the onset of the disease, but evidence suggests that chronic and low-grade inflammatory processes play an important role in their progression. Studies have identified similar signaling pathways and changes in resident immune cells within the retina among these diseases. Herein, we will discuss some of these studies and propose how understanding this inflammatory response could aid in the development of therapies.
Collapse
Affiliation(s)
- Michael T Massengill
- Department of Molecular Genetics & Microbiology, University of Florida College of Medicine, Gainesville, FL, USA
| | - Chulbul M Ahmed
- Department of Molecular Genetics & Microbiology, University of Florida College of Medicine, Gainesville, FL, USA
| | - Alfred S Lewin
- Department of Molecular Genetics & Microbiology, University of Florida College of Medicine, Gainesville, FL, USA
| | - Cristhian J Ildefonso
- Department of Ophthalmology, University of Florida College of Medicine, Gainesville, FL, USA.
| |
Collapse
|
68
|
Ling Y, Xiong F. Associations of TLR4 gene polymorphisms with the risk of age-related macular degeneration in a Chinese Han population. Medicine (Baltimore) 2019; 98:e15583. [PMID: 31083239 PMCID: PMC6531106 DOI: 10.1097/md.0000000000015583] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
The study was designed to reveal the relationship of toll-like receptor 4 (TLR4, rs1927914 and rs1927907) polymorphisms with risk of age-related macular degeneration (AMD), as well as the adjustment of this association by some environmental and lifestyle factors in Chinese Han population.TLR4 polymorphisms were genotyped by polymerase chain reaction-restricted fragment length polymorphisms and direct sequencing method in 138 AMD patients and 146 healthy controls. Genotype distribution in the control group was checked with Hardy-Weinberg equilibrium. Association of TLR4 polymorphisms and AMD risk was evaluated by χ test and adjusted by age and sex, smoking and drinking. Odds ratio (OR) with 95% confidence interval (95% CI) was used to represent the association strength. Logistic regressive analysis was used to calculate the adjusted OR values.CC genotype of rs1927914 had significantly lower frequency in AMD patients (P = .010), indicated a negative association with AMD risk (crude: OR = 0.358, 95% CI = 0.162-0.791; adjusted: OR = 0.355, 95% CI = 0.160-0.789). C allele of rs1927914 might decrease the susceptibility of AMD (crude: OR = 0.698, 95% CI = 0.497-0.982; adjusted: OR = 0.698, 95% CI = 0.495-0.984). No significant association has been discovered between TLR4 rs1927907 polymorphism and AMD susceptibility. Strong linkage disequilibrium existed between rs1927914 and rs1927907 polymorphisms. C-C haplotype was negatively associated with AMD risk (OR = 0.242, 95% CI = 0.121-0.485; OR = 0.242, 95% CI = 0.120-0.488).CC genotype and C allele of rs1927914 were significantly associated with the decreased AMD susceptibility.
Collapse
|
69
|
Fatty acids and oxidized lipoproteins contribute to autophagy and innate immunity responses upon the degeneration of retinal pigment epithelium and development of age-related macular degeneration. Biochimie 2019; 159:49-54. [DOI: 10.1016/j.biochi.2018.07.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 07/17/2018] [Indexed: 12/18/2022]
|
70
|
Noailles A, Kutsyr O, Maneu V, Ortuño-Lizarán I, Campello L, de Juan E, Gómez-Vicente V, Cuenca N, Lax P. The Absence of Toll-Like Receptor 4 Mildly Affects the Structure and Function in the Adult Mouse Retina. Front Cell Neurosci 2019; 13:59. [PMID: 30873007 PMCID: PMC6401850 DOI: 10.3389/fncel.2019.00059] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 02/05/2019] [Indexed: 12/26/2022] Open
Abstract
The innate immune Toll-like receptor (TLR) family plays essential roles in cell proliferation, survival and function of the central nervous system. However, the way in which TLRs contribute to the development and maintenance of proper retinal structure and function remains uncertain. In this work, we assess the effect of genetic TLR4 deletion on the morphology and function of the retina in mice. Visual acuity and retinal responsiveness were evaluated in TLR4 knockout and wild type C57BL/6J control mice by means of an optomotor test and electroretinography, respectively, from P20 to P360. Retinal structure was also analyzed in both strains using confocal and electron microscopy. ERG data showed impaired retinal responsiveness in TLR4 KO mice, in comparison to wild type animals. The amplitudes of the scotopic a-waves were less pronounced in TLR4-deficient mice than in wild-type animals from P30 to P360, and TLR4 KO mice presented scotopic b-wave amplitudes smaller than those of age-matched control mice at all ages studied (P20 to P360). Visual acuity was also relatively poorer in TLR4 KO as compared to C57BL/6J mice from P20 to P360, with significant differences at P30 and P60. Immunohistochemical analysis of retinal vertical sections showed no differences between TLR4 KO and C57BL/6J mice, in terms of either photoreceptor number or photoreceptor structure. Horizontal cells also demonstrated no morphological differences between TLR4 KO and wild-type mice. However, TLR4 KO mice exhibited a lower density of bipolar cells (15% less at P30) and thus fewer bipolar cell dendrites than the wild type control mouse, even though both confocal and electron microscopy images showed no morphologic abnormalities in the synaptic contacts between the photoreceptors and second order neurons. Microglial cell density was significantly lower (26% less at P30) in TLR4 KO mice as compared to wild-type control mice. These results suggest that TLR4 deletion causes functional alterations in terms of visual response and acuity, probably through the loss of bipolar cells and microglia, but this receptor is not essential for the processing of visual information in the retina.
Collapse
Affiliation(s)
- Agustina Noailles
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Oksana Kutsyr
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Victoria Maneu
- Department of Optics, Pharmacology and Anatomy, University of Alicante, Alicante, Spain
| | - Isabel Ortuño-Lizarán
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Laura Campello
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Emilio de Juan
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Violeta Gómez-Vicente
- Department of Optics, Pharmacology and Anatomy, University of Alicante, Alicante, Spain
| | - Nicolás Cuenca
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain.,Institute Ramón Margalef, University of Alicante, Alicante, Spain
| | - Pedro Lax
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| |
Collapse
|
71
|
Léveillard T, Philp NJ, Sennlaub F. Is Retinal Metabolic Dysfunction at the Center of the Pathogenesis of Age-related Macular Degeneration? Int J Mol Sci 2019; 20:ijms20030762. [PMID: 30754662 PMCID: PMC6387069 DOI: 10.3390/ijms20030762] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 02/04/2019] [Accepted: 02/05/2019] [Indexed: 01/12/2023] Open
Abstract
The retinal pigment epithelium (RPE) forms the outer blood⁻retina barrier and facilitates the transepithelial transport of glucose into the outer retina via GLUT1. Glucose is metabolized in photoreceptors via the tricarboxylic acid cycle (TCA) and oxidative phosphorylation (OXPHOS) but also by aerobic glycolysis to generate glycerol for the synthesis of phospholipids for the renewal of their outer segments. Aerobic glycolysis in the photoreceptors also leads to a high rate of production of lactate which is transported out of the subretinal space to the choroidal circulation by the RPE. Lactate taken up by the RPE is converted to pyruvate and metabolized via OXPHOS. Excess lactate in the RPE is transported across the basolateral membrane to the choroid. The uptake of glucose by cone photoreceptor cells is enhanced by rod-derived cone viability factor (RdCVF) secreted by rods and by insulin signaling. Together, the three cells act as symbiotes: the RPE supplies the glucose from the choroidal circulation to the photoreceptors, the rods help the cones, and both produce lactate to feed the RPE. In age-related macular degeneration this delicate ménage à trois is disturbed by the chronic infiltration of inflammatory macrophages. These immune cells also rely on aerobic glycolysis and compete for glucose and produce lactate. We here review the glucose metabolism in the homeostasis of the outer retina and in macrophages and hypothesize what happens when the metabolism of photoreceptors and the RPE is disturbed by chronic inflammation.
Collapse
Affiliation(s)
- Thierry Léveillard
- . Department of Genetics, Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France.
| | - Nancy J Philp
- . Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| | - Florian Sennlaub
- . Department of Therapeutics, Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France.
| |
Collapse
|
72
|
Kramer J, Chirco KR, Lamba DA. Immunological Considerations for Retinal Stem Cell Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1186:99-119. [PMID: 31654387 DOI: 10.1007/978-3-030-28471-8_4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
There is an increasing effort toward generating replacement cells for neuronal application due to the nonregenerative nature of these tissues. While much progress has been made toward developing methodologies to generate these cells, there have been limited improvements in functional restoration. Some of these are linked to the degenerative and often nonreceptive microenvironment that the new cells need to integrate into. In this chapter, we will focus on the status and role of the immune microenvironment of the retina during homeostasis and disease states. We will review changes in both innate and adaptive immunity as well as the role of immune rejection in stem cell replacement therapies. The chapter will end with a discussion of immune-modulatory strategies that have helped to ameliorate these effects and could potentially improve functional outcome for cell replacement therapies for the eye.
Collapse
Affiliation(s)
- Joshua Kramer
- Buck Institute for Research on Aging, Novato, CA, USA
| | | | - Deepak A Lamba
- Department of Ophthalmology, University of California San Francisco, San Francisco, CA, USA. .,Buck Institute for Research on Aging, Novato, CA, USA.
| |
Collapse
|
73
|
Saadane A, Mast N, Trichonas G, Chakraborty D, Hammer S, Busik JV, Grant MB, Pikuleva IA. Retinal Vascular Abnormalities and Microglia Activation in Mice with Deficiency in Cytochrome P450 46A1-Mediated Cholesterol Removal. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 189:405-425. [PMID: 30448403 DOI: 10.1016/j.ajpath.2018.10.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 09/27/2018] [Accepted: 10/15/2018] [Indexed: 12/11/2022]
Abstract
CYP46A1 is the cytochrome P450 enzyme that converts cholesterol to 24-hydroxycholesterol, a cholesterol elimination product and a potent liver X receptor (LXR) ligand. We conducted retinal characterizations of Cyp46a1-/- mice that had normal fasting blood glucose levels but up to a 1.8-fold increase in retinal cholesterol. The retina of Cyp46a1-/- mice exhibited venous beading and tortuosity, microglia/macrophage activation, and increased vascular permeability, features commonly associated with diabetic retinopathy. The expression of Lxrα and Lxrβ was increased in both the whole Cyp46a1-/- retina and retinal macroglia/macrophages. The LXR-target genes were affected as well, primarily in activated microglial cells and macrophages. In the latter, the LXR-transactivated genes (Abca1, Abcg1, Apod, Apoe, Mylip, and Arg2) were up-regulated; similarly, there was an up-regulation of the LXR-transrepressed genes (Ccl2, Ptgs2, Cxcl1, Il1b, Il6, Nos2, and Tnfa). For comparison, gene expression was investigated in bone marrow-derived macrophages from Cyp46a1-/- mice as well as retinal and bone marrow-derived macrophages from Cyp27a1-/- and Cyp27a1-/-Cyp46a1-/- mice. CYP46A1 expression was detected in retinal endothelial cells, and this expression was increased in the proinflammatory environment. Retinal Cyp46a1-/- phosphoproteome revealed altered phosphorylation of 30 different proteins, including tight junction protein zonula occludens 1 and aquaporin 4. Collectively, the data obtained establish metabolic and regulatory significance of CYP46A1 for the retina and suggest pharmacologic activation of CYP46A1 as a potential therapeutic approach to dyslipidemia-induced retinal damage.
Collapse
Affiliation(s)
- Aicha Saadane
- Department of Ophthalmology and Visual Sciences, the University Hospitals, Case Western Reserve University, Cleveland, Ohio
| | - Natalia Mast
- Department of Ophthalmology and Visual Sciences, the University Hospitals, Case Western Reserve University, Cleveland, Ohio
| | - George Trichonas
- Department of Ophthalmology and Visual Sciences, the University Hospitals, Case Western Reserve University, Cleveland, Ohio
| | | | - Sandra Hammer
- Department of Physiology, Michigan State University, East Lansing, Michigan
| | - Julia V Busik
- Department of Physiology, Michigan State University, East Lansing, Michigan
| | - Maria B Grant
- Department of Ophthalmology, University of Alabama, Birmingham, Alabama
| | - Irina A Pikuleva
- Department of Ophthalmology and Visual Sciences, the University Hospitals, Case Western Reserve University, Cleveland, Ohio.
| |
Collapse
|
74
|
Abstract
Microglia, the primary resident immune cell type, constitute a key population of glia in the retina. Recent evidence indicates that microglia play significant functional roles in the retina at different life stages. During development, retinal microglia regulate neuronal survival by exerting trophic influences and influencing programmed cell death. During adulthood, ramified microglia in the plexiform layers interact closely with synapses to maintain synaptic structure and function that underlie the retina's electrophysiological response to light. Under pathological conditions, retinal microglia participate in potentiating neurodegeneration in diseases such as glaucoma, retinitis pigmentosa, and age-related neurodegeneration by producing proinflammatory neurotoxic cytokines and removing living neurons via phagocytosis. Modulation of pathogenic microglial activation states and effector mechanisms has been linked to neuroprotection in animal models of retinal diseases. These findings have led to the design of early proof-of-concept clinical trials with microglial modulation as a therapeutic strategy.
Collapse
Affiliation(s)
- Sean M. Silverman
- Unit on Neuron-Glia Interactions in Retinal Disease, National Eye Institute, National Institutes of Health, Bethesda, Maryland 20892, USA;,
| | - Wai T. Wong
- Unit on Neuron-Glia Interactions in Retinal Disease, National Eye Institute, National Institutes of Health, Bethesda, Maryland 20892, USA;,
| |
Collapse
|
75
|
Palczewska G, Stremplewski P, Suh S, Alexander N, Salom D, Dong Z, Ruminski D, Choi EH, Sears AE, Kern TS, Wojtkowski M, Palczewski K. Two-photon imaging of the mammalian retina with ultrafast pulsing laser. JCI Insight 2018; 3:121555. [PMID: 30185665 DOI: 10.1172/jci.insight.121555] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 07/24/2018] [Indexed: 12/13/2022] Open
Abstract
Noninvasive imaging of visual system components in vivo is critical for understanding the causal mechanisms of retinal diseases and for developing therapies for their treatment. However, ultraviolet light needed to excite endogenous fluorophores that participate in metabolic processes of the retina is highly attenuated by the anterior segment of the human eye. In contrast, 2-photon excitation fluorescence imaging with pulsed infrared light overcomes this obstacle. Reducing retinal exposure to laser radiation remains a major barrier in advancing this technology to studies in humans. To increase fluorescence intensity and reduce the requisite laser power, we modulated ultrashort laser pulses with high-order dispersion compensation and applied sensorless adaptive optics and custom image recovery software and observed an over 300% increase in fluorescence of endogenous retinal fluorophores when laser pulses were shortened from 75 fs to 20 fs. No functional or structural changes to the retina were detected after exposure to 2-photon excitation imaging light with 20-fs pulses. Moreover, wide bandwidth associated with short pulses enables excitation of multiple fluorophores with different absorption spectra and thus can provide information about their relative changes and intracellular distribution. These data constitute a substantial advancement for safe 2-photon fluorescence imaging of the human eye.
Collapse
Affiliation(s)
| | - Patrycjusz Stremplewski
- Department of Physical Chemistry of Biological Systems, Institute of Physical Chemistry, Polish Academy of Sciences, Warsaw, Poland
| | - Susie Suh
- Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Nathan Alexander
- Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - David Salom
- Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Zhiqian Dong
- Polgenix, Inc., Department of Medical Devices, Cleveland, Ohio, USA
| | - Daniel Ruminski
- Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Elliot H Choi
- Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Avery E Sears
- Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Timothy S Kern
- Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Maciej Wojtkowski
- Department of Physical Chemistry of Biological Systems, Institute of Physical Chemistry, Polish Academy of Sciences, Warsaw, Poland
| | - Krzysztof Palczewski
- Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
76
|
Lorés-Motta L, Riaz M, Grunin M, Corominas J, van Asten F, Pauper M, Leenders M, Richardson AJ, Muether P, Cree AJ, Griffiths HL, Pham C, Belanger MC, Meester-Smoor MA, Ali M, Heid IM, Fritsche LG, Chakravarthy U, Gale R, McKibbin M, Inglehearn CF, Schlingemann RO, Omar A, Chen J, Koenekoop RK, Fauser S, Guymer RH, Hoyng CB, de Jong EK, Lotery AJ, Mitchell P, den Hollander AI, Baird PN, Chowers I. Association of Genetic Variants With Response to Anti-Vascular Endothelial Growth Factor Therapy in Age-Related Macular Degeneration. JAMA Ophthalmol 2018; 136:875-884. [PMID: 29852030 PMCID: PMC6142943 DOI: 10.1001/jamaophthalmol.2018.2019] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 04/02/2018] [Indexed: 02/04/2023]
Abstract
Importance Visual acuity (VA) outcomes differ considerably among patients with neovascular age-related macular degeneration (nAMD) treated with anti-vascular endothelial growth factor (VEGF) drugs. Identification of pharmacogenetic associations may help clinicians understand the mechanisms underlying this variability as well as pave the way for personalized treatment in nAMD. Objective To identify genetic factors associated with variability in the response to anti-VEGF therapy for patients with nAMD. Design, Setting, and Participants In this multicenter genome-wide association study, 678 patients with nAMD with genome-wide genotyping data were included in the discovery phase; 1380 additional patients with nAMD were genotyped for selected common variants in the replication phase. All participants received 3 monthly injections of bevacizumab or ranibizumab. Clinical data were evaluated for inclusion/exclusion criteria from October 2014 to October 2015, followed by data analysis from October 2015 to February 2016. For replication cohort genotyping, clinical data collection and analysis (including meta-analysis) was performed from March 2016 to April 2017. Main Outcomes and Measures Change in VA after the loading dose of 3 monthly anti-VEGF injections compared with baseline. Results Of the 2058 included patients, 1210 (58.8%) were women, and the mean (SD) age across all cohorts was 78 (7.4) years. Patients included in the discovery cohort and most of the patients in the replication cohorts were of European descent. The mean (SD) baseline VA was 51.3 (20.3) Early Treatment Diabetic Retinopathy Study (ETDRS) score letters, and the mean (SD) change in VA after the loading dose of 3 monthly injections was a gain of 5.1 (13.9) ETDRS score letters (ie, 1-line gain). Genome-wide single-variant analyses of common variants revealed 5 independent loci that reached a P value less than 10 × 10-5. After replication and meta-analysis of the lead variants, rs12138564 located in the CCT3 gene remained nominally associated with a better treatment outcome (ETDRS letter gain, 1.7; β, 0.034; SE, 0.008; P = 1.38 × 10-5). Genome-wide gene-based optimal unified sequence kernel association test of rare variants showed genome-wide significant associations for the C10orf88 (P = 4.22 × 10-7) and UNC93B1 (P = 6.09 × 10-7) genes, in both cases leading to a worse treatment outcome. Patients carrying rare variants in the C10orf88 and UNC93B1 genes lost a mean (SD) VA of 30.6 (17.4) ETDRS score letters (ie, loss of 6.09 lines) and 26.5 (13.8) ETDRS score letters (ie, loss of 5.29 lines), respectively, after 3 months of anti-VEGF treatment. Conclusions and Relevance We propose that there is a limited contribution of common genetic variants to variability in nAMD treatment response. Our results suggest that rare protein-altering variants in the C10orf88 and UNC93B1 genes are associated with a worse response to anti-VEGF therapy in patients with nAMD, but these results require further validation in other cohorts.
Collapse
Affiliation(s)
- Laura Lorés-Motta
- Department of Ophthalmology, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Moeen Riaz
- Centre for Eye Research Australia, Department of Surgery in Ophthalmology, University of Melbourne, Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia
- Public Health Genomics, School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Michelle Grunin
- Department of Ophthalmology, Hebrew University Hadassah Medical School, Hadassah Medical Center–Hebrew University, Jerusalem, Israel
| | - Jordi Corominas
- Department of Ophthalmology, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
- Department of Human Genetics, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Freekje van Asten
- Division of Epidemiology and Clinical Application, National Eye Institute, National Institutes of Health, Bethesda, Maryland
- Neurobiology, Neurodegeneration, and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Marc Pauper
- Department of Ophthalmology, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
- Department of Human Genetics, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Mathieu Leenders
- Department of Ophthalmology, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Andrea J. Richardson
- Centre for Eye Research Australia, Department of Surgery in Ophthalmology, University of Melbourne, Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia
| | - Philipp Muether
- Department of Ophthalmology, University Hospital of Cologne, Cologne, Germany
| | - Angela J. Cree
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, England
| | - Helen L. Griffiths
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, England
| | - Connie Pham
- Department Ophthalmology, McGill University Health Centre, Montreal, Québec, Canada
| | | | | | - Manir Ali
- Section of Ophthalmology and Neuroscience, Leeds Institute of Molecular Medicine, University of Leeds, Leeds, England
| | - Iris M. Heid
- Department of Genetic Epidemiology, University of Regensburg, Regensburg, Germany
| | - Lars G. Fritsche
- Norwegian University of Science and Technology, Trondheim, Norway
| | | | | | | | - Chris F. Inglehearn
- Section of Ophthalmology and Neuroscience, Leeds Institute of Molecular Medicine, University of Leeds, Leeds, England
- Eye Clinic, St James’s University Hospital, Leeds, England
| | - Reinier O. Schlingemann
- Department of Ophthalmology, Ocular Angiogenesis Group, Academic Medical Center, Amsterdam, the Netherlands
- Netherlands Institute for Neuroscience, Amsterdam, the Netherlands
| | - Amer Omar
- Montreal Retina Institute, Westmount, Québec, Canada
| | - John Chen
- Department of Pediatric Surgery, McGill University Health Centre, Montreal, Québec, Canada
- Department of Human Genetics, McGill University Health Centre, Montreal, Québec, Canada
- Department of Ophthalmology, McGill University Health Centre, Montreal, Québec, Canada
| | - Robert K. Koenekoop
- Department of Pediatric Surgery, McGill University Health Centre, Montreal, Québec, Canada
- Department of Human Genetics, McGill University Health Centre, Montreal, Québec, Canada
- Department of Ophthalmology, McGill University Health Centre, Montreal, Québec, Canada
| | - Sascha Fauser
- Department of Ophthalmology, University Hospital of Cologne, Cologne, Germany
- Roche Pharma Research and Early Development, Hoffmann–La Roche, Basel, Switzerland
| | - Robyn H. Guymer
- Centre for Eye Research Australia, Department of Surgery in Ophthalmology, University of Melbourne, Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia
| | - Carel B. Hoyng
- Department of Ophthalmology, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Eiko K. de Jong
- Department of Ophthalmology, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Andrew J. Lotery
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, England
| | - Paul Mitchell
- Centre for Vision Research, Department of Ophthalmology and Westmead Millennium Institute for Medical Research, University of Sydney, Sydney, New South Wales, Australia
| | - Anneke I. den Hollander
- Department of Ophthalmology, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
- Department of Human Genetics, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Paul N. Baird
- Centre for Eye Research Australia, Department of Surgery in Ophthalmology, University of Melbourne, Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia
| | - Itay Chowers
- Department of Ophthalmology, Hebrew University Hadassah Medical School, Hadassah Medical Center–Hebrew University, Jerusalem, Israel
| |
Collapse
|
77
|
Ayata P, Badimon A, Strasburger HJ, Duff MK, Montgomery SE, Loh YHE, Ebert A, Pimenova AA, Ramirez BR, Chan AT, Sullivan JM, Purushothaman I, Scarpa JR, Goate AM, Busslinger M, Shen L, Losic B, Schaefer A. Epigenetic regulation of brain region-specific microglia clearance activity. Nat Neurosci 2018; 21:1049-1060. [PMID: 30038282 PMCID: PMC6090564 DOI: 10.1038/s41593-018-0192-3] [Citation(s) in RCA: 293] [Impact Index Per Article: 48.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Accepted: 06/01/2018] [Indexed: 12/15/2022]
Abstract
The rapid elimination of dying neurons and nonfunctional synapses in the brain is carried out by microglia, the resident myeloid cells of the brain. Here we show that microglia clearance activity in the adult brain is regionally regulated and depends on the rate of neuronal attrition. Cerebellar, but not striatal or cortical, microglia exhibited high levels of basal clearance activity, which correlated with an elevated degree of cerebellar neuronal attrition. Exposing forebrain microglia to apoptotic cells activated gene-expression programs supporting clearance activity. We provide evidence that the polycomb repressive complex 2 (PRC2) epigenetically restricts the expression of genes that support clearance activity in striatal and cortical microglia. Loss of PRC2 leads to aberrant activation of a microglia clearance phenotype, which triggers changes in neuronal morphology and behavior. Our data highlight a key role of epigenetic mechanisms in preventing microglia-induced neuronal alterations that are frequently associated with neurodegenerative and psychiatric diseases.
Collapse
Affiliation(s)
- Pinar Ayata
- Fishberg Department of Neuroscience, Department of Psychiatry, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ana Badimon
- Fishberg Department of Neuroscience, Department of Psychiatry, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hayley J Strasburger
- Fishberg Department of Neuroscience, Department of Psychiatry, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mary Kaye Duff
- Fishberg Department of Neuroscience, Department of Psychiatry, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sarah E Montgomery
- Fishberg Department of Neuroscience, Department of Psychiatry, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yong-Hwee E Loh
- Fishberg Department of Neuroscience, Department of Psychiatry, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anja Ebert
- Research Institute of Molecular Pathology, Vienna Biocenter, Vienna, Austria
| | - Anna A Pimenova
- Fishberg Department of Neuroscience, Department of Psychiatry, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Brianna R Ramirez
- Fishberg Department of Neuroscience, Department of Psychiatry, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Andrew T Chan
- Fishberg Department of Neuroscience, Department of Psychiatry, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Josefa M Sullivan
- Fishberg Department of Neuroscience, Department of Psychiatry, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Immanuel Purushothaman
- Fishberg Department of Neuroscience, Department of Psychiatry, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Joseph R Scarpa
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alison M Goate
- Fishberg Department of Neuroscience, Department of Psychiatry, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Meinrad Busslinger
- Research Institute of Molecular Pathology, Vienna Biocenter, Vienna, Austria
| | - Li Shen
- Fishberg Department of Neuroscience, Department of Psychiatry, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bojan Losic
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Tisch Cancer Institute, Cancer Immunology Program, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anne Schaefer
- Fishberg Department of Neuroscience, Department of Psychiatry, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA. .,Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
78
|
Rashid K, Wolf A, Langmann T. Microglia Activation and Immunomodulatory Therapies for Retinal Degenerations. Front Cell Neurosci 2018; 12:176. [PMID: 29977192 PMCID: PMC6021747 DOI: 10.3389/fncel.2018.00176] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 06/05/2018] [Indexed: 01/05/2023] Open
Abstract
A chronic pro-inflammatory environment is a hallmark of retinal degenerative diseases and neurological disorders that affect vision. Inflammatory responses during retinal pathophysiology are orchestrated by microglial cells which constitute the resident immune cell population. Following activation, microglia cells lose their ramified protrusions, proliferate and rapidly migrate to the damaged areas and resolve tissue damage. However, sustained presence of tissue stress primes microglia to become overreactive and results in the excessive production of pro-inflammatory mediators that favor retinal degenerative changes. Consequently, interventions aimed at overriding microglial pro-inflammatory and pro-oxidative properties may attenuate photoreceptor demise and preserve retinal integrity. We highlight the positive effects of ligands for the translocator protein 18 kDa (TSPO) and the cytokine interferon beta (IFN-β) in modulating microgliosis during retinal pathologies and discuss their plausible mechanisms of action.
Collapse
Affiliation(s)
- Khalid Rashid
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, University of Cologne, Cologne, Germany
| | - Anne Wolf
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, University of Cologne, Cologne, Germany
| | - Thomas Langmann
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, University of Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| |
Collapse
|
79
|
Cell Death Pathways in Mutant Rhodopsin Rat Models Identifies Genotype-Specific Targets Controlling Retinal Degeneration. Mol Neurobiol 2018; 56:1637-1652. [PMID: 29911255 DOI: 10.1007/s12035-018-1192-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 06/08/2018] [Indexed: 12/24/2022]
Abstract
Retinitis pigmentosa (RP) is a group of inherited neurological disorders characterized by rod photoreceptor cell death, followed by secondary cone cell death leading to progressive blindness. Currently, there are no viable treatment options for RP. Due to incomplete knowledge of the molecular signaling pathways associated with RP pathogenesis, designing therapeutic strategies remains a challenge. In particular, preventing secondary cone photoreceptor cell loss is a key goal in designing potential therapies. In this study, we identified the main drivers of rod cell death and secondary cone loss in the transgenic S334ter rhodopsin rat model, tested the efficacy of specific cell death inhibitors on retinal function, and compared the effect of combining drugs to target multiple pathways in the S334ter and P23H rhodopsin rat models. The primary driver of early rod cell death in the S334ter model was a caspase-dependent process, whereas cone cell death occurred though RIP3-dependent necroptosis. In comparison, rod cell death in the P23H model was via necroptotic signaling, whereas cone cell loss occurred through inflammasome activation. Combination therapy of four drugs worked better than the individual drugs in the P23H model but not in the S334ter model. These differences imply that treatment modalities need to be tailored for each genotype. Taken together, our data demonstrate that rationally designed genotype-specific drug combinations will be an important requisite to effectively target primary rod cell loss and more importantly secondary cone survival.
Collapse
|
80
|
Damage-associated molecular pattern recognition is required for induction of retinal neuroprotective pathways in a sex-dependent manner. Sci Rep 2018; 8:9115. [PMID: 29904087 PMCID: PMC6002365 DOI: 10.1038/s41598-018-27479-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 06/04/2018] [Indexed: 12/29/2022] Open
Abstract
Retinal degeneration is a common cause of irreversible blindness and is caused by the death of retinal light-sensitive neurons called photoreceptors. At the onset of degeneration, stressed photoreceptors cause retinal glial cells to secrete neuroprotective factors that slow the pace of degeneration. Leukemia inhibitory factor (LIF) is one such factor that is required for endogenous neuroprotection. Photoreceptors are known to release signals of cellular stress, called damage-associated molecular patterns (DAMPs) early in degeneration, and we hypothesized that receptors for DAMPs or pattern recognition receptors (PRRs) play a key role in the induction of LIF and neuroprotective stress responses in retinal glial cells. Toll-like receptor 2 (TLR2) is a well-established DAMP receptor. In our experiments, activation of TLR2 protected both male and female mice from light damage, while the loss of TLR2 in female mice did not impact photoreceptor survival. In contrast, induction of protective stress responses, microglial phenotype and photoreceptor survival were strongly impacted in male TLR2−/− mice. Lastly, using publicly available gene expression data, we show that TLR2 is expressed highly in resting microglia prior to injury, but is also induced in Müller cells in inherited retinal degeneration.
Collapse
|
81
|
Ozal SA, Turkekul K, Gurlu V, Guclu H, Erdogan S. Esculetin Protects Human Retinal Pigment Epithelial Cells from Lipopolysaccharide-induced Inflammation and Cell Death. Curr Eye Res 2018; 43:1169-1176. [DOI: 10.1080/02713683.2018.1481517] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- S. Altan Ozal
- Department of Ophthalmology, School of Medicine, Trakya University, Edirne, Turkey
| | - Kader Turkekul
- Department of Medical Biology, School of Medicine, Trakya University, Edirne, Turkey
| | - Vuslat Gurlu
- Department of Ophthalmology, School of Medicine, Trakya University, Edirne, Turkey
| | - Hande Guclu
- Department of Ophthalmology, School of Medicine, Trakya University, Edirne, Turkey
| | - Suat Erdogan
- Department of Medical Biology, School of Medicine, Trakya University, Edirne, Turkey
| |
Collapse
|
82
|
Rathnasamy G, Foulds WS, Ling EA, Kaur C. Retinal microglia - A key player in healthy and diseased retina. Prog Neurobiol 2018; 173:18-40. [PMID: 29864456 DOI: 10.1016/j.pneurobio.2018.05.006] [Citation(s) in RCA: 139] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 03/09/2018] [Accepted: 05/29/2018] [Indexed: 01/04/2023]
Abstract
Microglia, the resident immune cells of the brain and retina, are constantly engaged in the surveillance of their surrounding neural tissue. During embryonic development they infiltrate the retinal tissues and participate in the phagocytosis of redundant neurons. The contribution of microglia in maintaining the purposeful and functional histo-architecture of the adult retina is indispensable. Within the retinal microenvironment, robust microglial activation is elicited by subtle changes caused by extrinsic and intrinsic factors. When there is a disturbance in the cell-cell communication between microglia and other retinal cells, for example in retinal injury, the activated microglia can manifest actions that can be detrimental. This is evidenced by activated microglia secreting inflammatory mediators that can further aggravate the retinal injury. Microglial activation as a harbinger of a variety of retinal diseases is well documented by many studies. In addition, a change in the microglial phenotype which may be associated with aging, may predispose the retina to age-related diseases. In light of the above, the focus of this review is to highlight the role played by microglia in the healthy and diseased retina, based on findings of our own work and from that of others.
Collapse
Affiliation(s)
- Gurugirijha Rathnasamy
- Department of Anatomy, Yong Loo Lin School of Medicine, Blk MD10, 4 Medical Drive, National University of Singapore, 117594, Singapore; Department of Ophthalmology and Visual Sciences, School of Medicine and Public Health, University of Wisconsin, Madison, WI, 53706, United States
| | - Wallace S Foulds
- Singapore Eye Research Institute Level 6, The Academia, Discovery Tower, 20 College Road, 169856, Singapore; University of Glasgow, Glasgow, Scotland, G12 8QQ, United Kingdom
| | - Eng-Ang Ling
- Department of Anatomy, Yong Loo Lin School of Medicine, Blk MD10, 4 Medical Drive, National University of Singapore, 117594, Singapore
| | - Charanjit Kaur
- Department of Anatomy, Yong Loo Lin School of Medicine, Blk MD10, 4 Medical Drive, National University of Singapore, 117594, Singapore.
| |
Collapse
|
83
|
Dejos C, Kuny S, Han WH, Capel H, Lemieux H, Sauvé Y. Photoreceptor-induced RPE phagolysosomal maturation defects in Stargardt-like Maculopathy (STGD3). Sci Rep 2018; 8:5944. [PMID: 29654292 PMCID: PMC5899129 DOI: 10.1038/s41598-018-24357-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 03/28/2018] [Indexed: 11/09/2022] Open
Abstract
For many neurodegenerative disorders, expression of a pathological protein by one cell type impedes function of other cell types, which in turn contributes to the death of the first cell type. In transgenic mice modelling Stargardt-like (STGD3) maculopathy, human mutant ELOVL4 expression by photoreceptors is associated with defects in the underlying retinal pigment epithelium (RPE). To examine how photoreceptors exert cytotoxic effects on RPE cells, transgenic ELOVL4 (TG1-2 line; TG) and wild-type (WT) littermates were studied one month prior (preclinical stage) to onset of photoreceptor loss (two months). TG photoreceptor outer segments presented to human RPE cells are recognized and internalized into phagosomes, but their digestion is delayed. Live RPE cell imaging pinpoints decreased numbers of acidified phagolysomes. In vivo, master regulator of lysosomal genes, transcription factor EB (TFEB), and key lysosomal enzyme Cathepsin D are both unaffected. Oxidative stress, as ruled out with high-resolution respirometry, does not play a role at such an early stage. Upregulation of CRYBA1/A3 and phagocytic cells (microglia/macrophages) interposed between RPE and photoreceptors support adaptive responses to processing delays. Impaired phagolysosomal maturation is observed in RPE of mice expressing human mutant ELOVL4 in their photoreceptors prior to photoreceptor death and associated vision loss.
Collapse
Affiliation(s)
- Camille Dejos
- Department of Ophthalmology and Visual Sciences, University of Alberta, 7-45 Medical Sciences Building, Edmonton, AB, T6G 2H7, Canada
| | - Sharee Kuny
- Department of Ophthalmology and Visual Sciences, University of Alberta, 7-45 Medical Sciences Building, Edmonton, AB, T6G 2H7, Canada
| | - Woo Hyun Han
- Department of Ophthalmology and Visual Sciences, University of Alberta, 7-45 Medical Sciences Building, Edmonton, AB, T6G 2H7, Canada
| | - Heather Capel
- Department of Physiology, University of Alberta, 7-45 Medical Sciences Building, Edmonton, AB, T6G 2H7, Canada
| | - Hélène Lemieux
- Faculty Saint-Jean, University of Alberta, 8406 Rue Marie-Anne Gaboury Northwest, Edmonton, AB, T6C 4G9, Canada
| | - Yves Sauvé
- Department of Ophthalmology and Visual Sciences, University of Alberta, 7-45 Medical Sciences Building, Edmonton, AB, T6G 2H7, Canada. .,Department of Physiology, University of Alberta, 7-45 Medical Sciences Building, Edmonton, AB, T6G 2H7, Canada.
| |
Collapse
|
84
|
Parmar T, Parmar VM, Perusek L, Georges A, Takahashi M, Crabb JW, Maeda A. Lipocalin 2 Plays an Important Role in Regulating Inflammation in Retinal Degeneration. THE JOURNAL OF IMMUNOLOGY 2018; 200:3128-3141. [PMID: 29602770 DOI: 10.4049/jimmunol.1701573] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 03/05/2018] [Indexed: 12/17/2022]
Abstract
It has become increasingly important to understand how retinal inflammation is regulated because inflammation plays a role in retinal degenerative diseases. Lipocalin 2 (LCN2), an acute stress response protein with multiple innate immune functions, is increased in ATP-binding cassette subfamily A member 4 (Abca4) -/- retinol dehydrogenase 8 (Rdh8) -/- double-knockout mice, an animal model for Stargardt disease and age-related macular degeneration (AMD). To examine roles of LCN2 in retinal inflammation and degeneration, Lcn2-/-Abca4-/-Rdh8-/- triple-knockout mice were generated. Exacerbated inflammation following light exposure was observed in Lcn2-/-Abca4-/-Rdh8-/- mice as compared with Abca4-/-Rdh8-/- mice, with upregulation of proinflammatory genes and microglial activation. RNA array analyses revealed an increase in immune response molecules such as Ccl8, Ccl2, and Cxcl10 To further probe a possible regulatory role for LCN2 in retinal inflammation, we examined the in vitro effects of LCN2 on NF-κB signaling in human retinal pigmented epithelial (RPE) cells differentiated from induced pluripotent stem cells derived from healthy donors. We found that LCN2 induced expression of antioxidant enzymes heme oxygenase 1 and superoxide dismutase 2 in these RPE cells and could inhibit the cytotoxic effects of H2O2 and LPS. ELISA revealed increased LCN2 levels in plasma of patients with Stargardt disease, retinitis pigmentosa, and age-related macular degeneration as compared with healthy controls. Finally, overexpression of LCN2 in RPE cells displayed protection from cell death. Overall these results suggest that LCN2 is involved in prosurvival responses during cell stress and plays an important role in regulating inflammation during retinal degeneration.
Collapse
Affiliation(s)
- Tanu Parmar
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH 44106
| | - Vipul M Parmar
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH 44106
| | - Lindsay Perusek
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH 44106
| | - Anouk Georges
- Laboratory for Retinal Regeneration, RIKEN Center for Developmental Biology, Kobe, Hyogo 650-0047, Japan
| | - Masayo Takahashi
- Laboratory for Retinal Regeneration, RIKEN Center for Developmental Biology, Kobe, Hyogo 650-0047, Japan
| | - John W Crabb
- Cole Eye Institute, Cleveland Clinic, OH 44195; and
| | - Akiko Maeda
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH 44106; .,Laboratory for Retinal Regeneration, RIKEN Center for Developmental Biology, Kobe, Hyogo 650-0047, Japan.,Department of Pharmacology, Case Western Reserve University, Cleveland, OH 44106
| |
Collapse
|
85
|
Taylor AW, Ng TF. Negative regulators that mediate ocular immune privilege. J Leukoc Biol 2018; 103:1179-1187. [PMID: 29431864 PMCID: PMC6240388 DOI: 10.1002/jlb.3mir0817-337r] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 12/15/2017] [Accepted: 01/10/2018] [Indexed: 08/13/2023] Open
Abstract
The ocular microenvironment has adapted several negative regulators of inflammation to maintain immune privilege and health of the visual axis. Several constitutively produced negative regulators within the eye TGF-β2, α-melanocyte stimulating hormone (α-MSH), Fas ligand (FasL), and PD-L1 standout because of their capacity to influence multiple pathways of inflammation, and that they are part of promoting immune tolerance. These regulators demonstrate the capacity of immune privilege to prevent the activation of inflammation, and to suppress activation of effector immune cells even under conditions of ocular inflammation induced by endotoxin and autoimmune disease. In addition, these negative regulators promote and expand immune cells that mediate regulatory and tolerogenic immunity. This in turn makes the immune cells themselves negative regulators of inflammation. This provides for a greater understanding of immune privilege in that it includes both molecular and cellular negative regulators of inflammation. This would mean that potentially new approaches to the treatment of autoimmune disease can be developed through the use of molecules and cells as negative regulators of inflammation.
Collapse
Affiliation(s)
- Andrew W Taylor
- Boston University School of Medicine, Boston, Massachusetts, USA
| | - Tat Fong Ng
- Boston University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
86
|
Retinal neuroinflammatory induced neuronal degeneration - Role of toll-like receptor-4 and relationship with gliosis. Exp Eye Res 2018; 169:99-110. [PMID: 29425879 DOI: 10.1016/j.exer.2018.02.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 01/21/2018] [Accepted: 02/03/2018] [Indexed: 12/18/2022]
Abstract
The purpose of this study was to explore retina-intrinsic neuroinflammatory reactions, effects on neuronal survival, relationship with classic gliosis, and possible role of the toll-like receptor 4 (TLR4). To isolate the adult retina from the systemic immune system, a previously described large animal explant culture model was used in which full-thickness porcine retinal sheets can be kept in vitro for extended time periods. Explants were kept for 5 days in vitro (DIV) and were treated with either; lipopolysaccharide (LPS), a Toll-like receptor-4 (TLR4) inhibitor (CLI-095), LPS + CLI-095, or solvent vehicle throughout the culture period after which retinal sections were examined with hematoxylin and eosin staining and extensive immunohistochemistry. In addition, culture medium from all explant groups was assayed for a panel of cytokines at 2 and 5DIV. Compared with in vivo controls, vehicle controls (CT) as well as CLI-095 explants displayed moderate reduction of total thickness and number of retinal neurons with upregulation of glial fibrillary acidic protein (GFAP) throughout the Müller cells. In contrast, LPS and LPS + CLI-095 treated counterparts showed extensive overall thinning with widespread neuronal degeneration but only minimal signs of classical Müller cell gliosis (limited upregulation of GFAP and no downregulation of glutamine synthetase (GS). These specimens also displayed a significantly increased expression of galectin-3 and TGF-beta activated kinase 1 (TAK1). Multiplex proteomic analysis of culture medium at 2DIV revealed elevated levels of IL-1β, IL-6, IL-4 and IL-12 in LPS-treated explants compared to CLI-095 and CT counterparts. LPS stimulation of the isolated adult retina results in substantial neuronal cell death despite only minimal signs of gliosis indicating a retina-intrinsic neuroinflammatory response directly related to the degenerative process. This response is characterized by early upregulation of several inflammatory related cytokines with subsequent upregulation of Galectin-3, TLR4 and TAK1. Pharmacological block of TLR4 does not attenuate neuronal loss indicating that LPS induced retinal degeneration is mediated by TLR4 independent neuroinflammatory pathways.
Collapse
|
87
|
Madeira MH, Rashid K, Ambrósio AF, Santiago AR, Langmann T. Blockade of microglial adenosine A2A receptor impacts inflammatory mechanisms, reduces ARPE-19 cell dysfunction and prevents photoreceptor loss in vitro. Sci Rep 2018; 8:2272. [PMID: 29396515 PMCID: PMC5797099 DOI: 10.1038/s41598-018-20733-2] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 01/22/2018] [Indexed: 01/22/2023] Open
Abstract
Age-related macular degeneration (AMD) is characterized by pathological changes in the retinal pigment epithelium (RPE) and loss of photoreceptors. Growing evidence has demonstrated that reactive microglial cells trigger RPE dysfunction and loss of photoreceptors, and inflammasome pathways and complement activation contribute to AMD pathogenesis. We and others have previously shown that adenosine A2A receptor (A2AR) blockade prevents microglia-mediated neuroinflammatory processes and mediates protection to the retina. However, it is still unknown whether blocking A2AR in microglia protects against the pathological features of AMD. Herein, we show that an A2AR antagonist, SCH58261, prevents the upregulation of the expression of pro-inflammatory mediators and the alterations in the complement system triggered by an inflammatory challenge in human microglial cells. Furthermore, blockade of A2AR in microglia decreases the inflammatory response, as well as complement and inflammasome activation, in ARPE-19 cells exposed to conditioned medium of activated microglia. Finally, we also show that blocking A2AR in human microglia increases the clearance of apoptotic photoreceptors. This study opens the possibility of using selective A2AR antagonists in therapy for AMD, by modulating the interplay between microglia, RPE and photoreceptors.
Collapse
Affiliation(s)
- M H Madeira
- Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal.,Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, University of Cologne, Cologne, Germany
| | - K Rashid
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, University of Cologne, Cologne, Germany
| | - A F Ambrósio
- Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal.,Association for Innovation and Biomedical Research on Light and Image (AIBILI), Coimbra, Portugal
| | - A R Santiago
- Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal.,Association for Innovation and Biomedical Research on Light and Image (AIBILI), Coimbra, Portugal
| | - T Langmann
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, University of Cologne, Cologne, Germany.
| |
Collapse
|
88
|
Mansour AM, Sheheitli H, Kucukerdonmez C, Sisk RA, Moura R, Moschos MM, Lima LH, Al-Shaar L, Arevalo JF, Maia M, Foster RE, Kayikcioglu O, Kozak I, Kurup S, Zegarra H, Gallego-Pinazo R, Hamam RN, Bejjani RA, Cinar E, Erakgün ET, Kimura A, Teixeira A. INTRAVITREAL DEXAMETHASONE IMPLANT IN RETINITIS PIGMENTOSA–RELATED CYSTOID MACULAR EDEMA. Retina 2018; 38:416-423. [DOI: 10.1097/iae.0000000000001542] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
89
|
Schur RM, Gao S, Yu G, Chen Y, Maeda A, Palczewski K, Lu ZR. New GABA modulators protect photoreceptor cells from light-induced degeneration in mouse models. FASEB J 2018; 32:3289-3300. [PMID: 29401616 DOI: 10.1096/fj.201701250r] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
No clinically approved therapies are currently available that prevent the onset of photoreceptor death in retinal degeneration. Signaling between retinal neurons is regulated by the release and uptake of neurotransmitters, wherein GABA is the main inhibitory neurotransmitter. In this work, novel 3-chloropropiophenone derivatives and the clinical anticonvulsants tiagabine and vigabatrin were tested to modulate GABA signaling and protect against light-induced retinal degeneration. Abca4-/-Rdh8-/- mice, an accelerated model of retinal degeneration, were exposed to intense light after prophylactic injections of one of these compounds. Imaging and functional assessments of the retina indicated that these compounds successfully protected photoreceptor cells from degeneration to maintain a full-visual-field response. Furthermore, these compounds demonstrated a strong safety profile in wild-type mice and did not compromise visual function or damage the retina, despite repeated administration. These results indicate that modulating inhibitory GABA signaling can offer prophylactic protection against light-induced retinal degeneration.-Schur, R. M., Gao, S., Yu, G., Chen, Y., Maeda, A., Palczewski, K., Lu, Z.-R. New GABA modulators protect photoreceptor cells from light-induced degeneration in mouse models.
Collapse
Affiliation(s)
- Rebecca M Schur
- Case Center for Biomolecular Engineering, Department of Biomedical Engineering, School of Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Songqi Gao
- Department of Pharmacology, Cleveland Center for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Guanping Yu
- Case Center for Biomolecular Engineering, Department of Biomedical Engineering, School of Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Yu Chen
- Department of Pharmacology, Cleveland Center for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Akiko Maeda
- Department of Ophthalmology and Visual Sciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Krzysztof Palczewski
- Department of Pharmacology, Cleveland Center for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Zheng-Rong Lu
- Case Center for Biomolecular Engineering, Department of Biomedical Engineering, School of Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
90
|
Sene A, Apte RS. Inflammation-Induced Photoreceptor Cell Death. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1074:203-208. [PMID: 29721945 DOI: 10.1007/978-3-319-75402-4_25] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Neuroinflammation is an important aspect of many diseases of the eye, and experimental animal models have been widely used to determine its impact on retinal homeostasis and neuron survival. Physical separation of the neurosensory retina from the underlying retinal pigment epithelium (RPE) results in activation and infiltration of macrophages. Numerous studies have shown the critical role of macrophages in retinal disease processes. In retinal detachment, accumulation of macrophages in the subretinal space is associated with changes in cytokine and chemokine profile which lead to photoreceptor cell death. Targeted disruption of macrophage chemotaxis significantly reduces retinal detachment-induced photoreceptor degeneration. Apoptosis is the predominant mechanism of cell death; however regulated necrosis is also a contributor of photoreceptor loss. Therefore, effective neuroprotective approaches could integrate combined inhibition of both apoptotic and regulated necrosis pathways.
Collapse
Affiliation(s)
- Abdoulaye Sene
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA.
| | - Rajendra S Apte
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA.,Department of Developmental Biology and Medicine, Washington University School of Medicine, St. Louis, MO, USA.,Department of Biology, Allergan, Inc., Irvine, CA, USA
| |
Collapse
|
91
|
Davies JMS, Cillard J, Friguet B, Cadenas E, Cadet J, Cayce R, Fishmann A, Liao D, Bulteau AL, Derbré F, Rébillard A, Burstein S, Hirsch E, Kloner RA, Jakowec M, Petzinger G, Sauce D, Sennlaub F, Limon I, Ursini F, Maiorino M, Economides C, Pike CJ, Cohen P, Salvayre AN, Halliday MR, Lundquist AJ, Jakowec NA, Mechta-Grigoriou F, Mericskay M, Mariani J, Li Z, Huang D, Grant E, Forman HJ, Finch CE, Sun PY, Pomatto LCD, Agbulut O, Warburton D, Neri C, Rouis M, Cillard P, Capeau J, Rosenbaum J, Davies KJA. The Oxygen Paradox, the French Paradox, and age-related diseases. GeroScience 2017; 39:499-550. [PMID: 29270905 PMCID: PMC5745211 DOI: 10.1007/s11357-017-0002-y] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 11/09/2017] [Indexed: 02/06/2023] Open
Abstract
A paradox is a seemingly absurd or impossible concept, proposition, or theory that is often difficult to understand or explain, sometimes apparently self-contradictory, and yet ultimately correct or true. How is it possible, for example, that oxygen "a toxic environmental poison" could be also indispensable for life (Beckman and Ames Physiol Rev 78(2):547-81, 1998; Stadtman and Berlett Chem Res Toxicol 10(5):485-94, 1997)?: the so-called Oxygen Paradox (Davies and Ursini 1995; Davies Biochem Soc Symp 61:1-31, 1995). How can French people apparently disregard the rule that high dietary intakes of cholesterol and saturated fats (e.g., cheese and paté) will result in an early death from cardiovascular diseases (Renaud and de Lorgeril Lancet 339(8808):1523-6, 1992; Catalgol et al. Front Pharmacol 3:141, 2012; Eisenberg et al. Nat Med 22(12):1428-1438, 2016)?: the so-called, French Paradox. Doubtless, the truth is not a duality and epistemological bias probably generates apparently self-contradictory conclusions. Perhaps nowhere in biology are there so many apparently contradictory views, and even experimental results, affecting human physiology and pathology as in the fields of free radicals and oxidative stress, antioxidants, foods and drinks, and dietary recommendations; this is particularly true when issues such as disease-susceptibility or avoidance, "healthspan," "lifespan," and ageing are involved. Consider, for example, the apparently paradoxical observation that treatment with low doses of a substance that is toxic at high concentrations may actually induce transient adaptations that protect against a subsequent exposure to the same (or similar) toxin. This particular paradox is now mechanistically explained as "Adaptive Homeostasis" (Davies Mol Asp Med 49:1-7, 2016; Pomatto et al. 2017a; Lomeli et al. Clin Sci (Lond) 131(21):2573-2599, 2017; Pomatto and Davies 2017); the non-damaging process by which an apparent toxicant can activate biological signal transduction pathways to increase expression of protective genes, by mechanisms that are completely different from those by which the same agent induces toxicity at high concentrations. In this review, we explore the influences and effects of paradoxes such as the Oxygen Paradox and the French Paradox on the etiology, progression, and outcomes of many of the major human age-related diseases, as well as the basic biological phenomenon of ageing itself.
Collapse
Affiliation(s)
- Joanna M S Davies
- The Medical Group, Internal Medicine, Rheumatology & Osteoporosis, Dermatology, Pulmonology, Ophthalmology, and Cardiology; the Hospital of the Good Samaritan, Los Angeles, CA, 90017, USA
- Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology Center, University of Southern California, Los Angeles, CA, 90089-0191, USA
| | - Josiane Cillard
- Lab de Biologie Cellulaire et Végétale, Faculté de Pharmacie, Université de Rennes, 35043, Rennes Cedex, France
| | - Bertrand Friguet
- Institut de Biologie Paris-Seine (IBPS), UMR CNRS 8256, Biological Adaptation and Ageing, Sorbonne Universités, UPMC Univ Paris 06, 75005, Paris, France
- INSERM ERL U1164, 75005, Paris, France
| | - Enrique Cadenas
- Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology Center, University of Southern California, Los Angeles, CA, 90089-0191, USA
- School of Pharmacy, University of Southern California, Los Angeles, CA, 90089-9121, USA
- Department of Biochemistry & Molecular Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, 90033, USA
| | - Jean Cadet
- Département de Médecine nucléaire et Radiobiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Québec, J1H 5N4, Canada
| | - Rachael Cayce
- The Medical Group, Internal Medicine, Rheumatology & Osteoporosis, Dermatology, Pulmonology, Ophthalmology, and Cardiology; the Hospital of the Good Samaritan, Los Angeles, CA, 90017, USA
| | - Andrew Fishmann
- The Medical Group, Internal Medicine, Rheumatology & Osteoporosis, Dermatology, Pulmonology, Ophthalmology, and Cardiology; the Hospital of the Good Samaritan, Los Angeles, CA, 90017, USA
| | - David Liao
- The Medical Group, Internal Medicine, Rheumatology & Osteoporosis, Dermatology, Pulmonology, Ophthalmology, and Cardiology; the Hospital of the Good Samaritan, Los Angeles, CA, 90017, USA
| | - Anne-Laure Bulteau
- Institut de Génomique Fonctionnelle de Lyon,ENS de Lyon, CNRS, 69364, Lyon Cedex 07, France
| | - Frédéric Derbré
- Laboratory for Movement, Sport and Health Sciences-EA 1274, M2S, Université de Rennes 2-ENS, Bruz, 35170, Rennes, France
| | - Amélie Rébillard
- Laboratory for Movement, Sport and Health Sciences-EA 1274, M2S, Université de Rennes 2-ENS, Bruz, 35170, Rennes, France
| | - Steven Burstein
- The Medical Group, Internal Medicine, Rheumatology & Osteoporosis, Dermatology, Pulmonology, Ophthalmology, and Cardiology; the Hospital of the Good Samaritan, Los Angeles, CA, 90017, USA
| | - Etienne Hirsch
- INSERM UMR 1127-CNRS UMR 7225, Institut du cerveau et de la moelle épinière-ICM Thérapeutique Expérimentale de la Maladie de Parkinson, Université Pierre et Marie Curie, 75651, Paris Cedex 13, France
| | - Robert A Kloner
- Huntington Medical Research Institutes, Pasadena, CA, 91105, USA
| | - Michael Jakowec
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Giselle Petzinger
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Delphine Sauce
- Chronic infections and Immune ageing, INSERM U1135, Hopital Pitie-Salpetriere, Pierre et Marie Curie University, 75013, Paris, France
| | | | - Isabelle Limon
- Institut de Biologie Paris-Seine (IBPS), UMR CNRS 8256, Biological Adaptation and Ageing, Sorbonne Universités, UPMC Univ Paris 06, 75005, Paris, France
| | - Fulvio Ursini
- Department of Molecular Medicine, University of Padova, 35121, Padova, Italy
| | - Matilde Maiorino
- Department of Molecular Medicine, University of Padova, 35121, Padova, Italy
| | - Christina Economides
- Los Angeles Cardiology Associates, Hospital of the Good Samaritan, Los Angeles, CA, 90017, USA
| | - Christian J Pike
- Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology Center, University of Southern California, Los Angeles, CA, 90089-0191, USA
- Division of Neurobiology, Department of Biological Sciences of the Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, CA, 90089-0191, USA
| | - Pinchas Cohen
- Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology Center, University of Southern California, Los Angeles, CA, 90089-0191, USA
- Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, 90033, USA
| | - Anne Negre Salvayre
- Lipid peroxidation, Signalling and Vascular Diseases INSERM U1048, 31432, Toulouse Cedex 4, France
| | - Matthew R Halliday
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Adam J Lundquist
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Nicolaus A Jakowec
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | | | - Mathias Mericskay
- Laboratoire de Signalisation et Physiopathologie Cardiovasculaire-Inserm UMR-S 1180, Faculté de Pharmacie, Université Paris-Sud, 92296 Châtenay-Malabry, Paris, France
| | - Jean Mariani
- Institut de Biologie Paris-Seine (IBPS), UMR CNRS 8256, Biological Adaptation and Ageing, Sorbonne Universités, UPMC Univ Paris 06, 75005, Paris, France
| | - Zhenlin Li
- Institut de Biologie Paris-Seine (IBPS), UMR CNRS 8256, Biological Adaptation and Ageing, Sorbonne Universités, UPMC Univ Paris 06, 75005, Paris, France
- INSERM ERL U1164, 75005, Paris, France
| | - David Huang
- Department of Radiation Oncology, Hospital of the Good Samaritan, Los Angeles, CA, 90017, USA
| | - Ellsworth Grant
- Department of Oncology & Hematology, Hospital of the Good Samaritan, Los Angeles, CA, 90017, USA
| | - Henry J Forman
- Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology Center, University of Southern California, Los Angeles, CA, 90089-0191, USA
| | - Caleb E Finch
- Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology Center, University of Southern California, Los Angeles, CA, 90089-0191, USA
- Los Angeles Cardiology Associates, Hospital of the Good Samaritan, Los Angeles, CA, 90017, USA
- Division of Molecular & Computational Biology, Department of Biological Sciences of the Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, CA, 90089-0191, USA
| | - Patrick Y Sun
- Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology Center, University of Southern California, Los Angeles, CA, 90089-0191, USA
- Division of Molecular & Computational Biology, Department of Biological Sciences of the Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, CA, 90089-0191, USA
| | - Laura C D Pomatto
- Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology Center, University of Southern California, Los Angeles, CA, 90089-0191, USA
- Division of Molecular & Computational Biology, Department of Biological Sciences of the Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, CA, 90089-0191, USA
| | - Onnik Agbulut
- Institut de Biologie Paris-Seine (IBPS), UMR CNRS 8256, Biological Adaptation and Ageing, Sorbonne Universités, UPMC Univ Paris 06, 75005, Paris, France
| | - David Warburton
- Children's Hospital of Los Angeles, Developmental Biology, Regenerative Medicine and Stem Cell Therapeutics program and the Center for Environmental Impact on Global Health Across the Lifespan at The Saban Research Institute, Los Angeles, CA, 90027, USA
- Department of Pediatrics, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, 90033, USA
| | - Christian Neri
- Institut de Biologie Paris-Seine (IBPS), UMR CNRS 8256, Biological Adaptation and Ageing, Sorbonne Universités, UPMC Univ Paris 06, 75005, Paris, France
| | - Mustapha Rouis
- Institut de Biologie Paris-Seine (IBPS), UMR CNRS 8256, Biological Adaptation and Ageing, Sorbonne Universités, UPMC Univ Paris 06, 75005, Paris, France
- INSERM ERL U1164, 75005, Paris, France
| | - Pierre Cillard
- Lab de Biologie Cellulaire et Végétale, Faculté de Pharmacie, Université de Rennes, 35043, Rennes Cedex, France
| | - Jacqueline Capeau
- DR Saint-Antoine UMR_S938, UPMC, Inserm Faculté de Médecine, Université Pierre et Marie Curie, 75012, Paris, France
| | - Jean Rosenbaum
- Scientific Service of the Embassy of France in the USA, Consulate General of France in Los Angeles, Los Angeles, CA, 90025, USA
| | - Kelvin J A Davies
- Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology Center, University of Southern California, Los Angeles, CA, 90089-0191, USA.
- Department of Biochemistry & Molecular Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, 90033, USA.
- Division of Molecular & Computational Biology, Department of Biological Sciences of the Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, CA, 90089-0191, USA.
| |
Collapse
|
92
|
On phagocytes and macular degeneration. Prog Retin Eye Res 2017; 61:98-128. [DOI: 10.1016/j.preteyeres.2017.06.002] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 05/29/2017] [Accepted: 06/05/2017] [Indexed: 12/17/2022]
|
93
|
Shinozaki Y, Kashiwagi K, Namekata K, Takeda A, Ohno N, Robaye B, Harada T, Iwata T, Koizumi S. Purinergic dysregulation causes hypertensive glaucoma-like optic neuropathy. JCI Insight 2017; 2:93456. [PMID: 28978804 DOI: 10.1172/jci.insight.93456] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 08/24/2017] [Indexed: 12/24/2022] Open
Abstract
Glaucoma is an optic neuropathy characterized by progressive degeneration of retinal ganglion cells (RGCs) and visual loss. Although one of the highest risk factors for glaucoma is elevated intraocular pressure (IOP) and reduction in IOP is the only proven treatment, the mechanism of IOP regulation is poorly understood. We report that the P2Y6 receptor is critical for lowering IOP and that ablation of the P2Y6 gene in mice (P2Y6KO) results in hypertensive glaucoma-like optic neuropathy. Topically applied uridine diphosphate, an endogenous selective agonist for the P2Y6 receptor, decreases IOP. The P2Y6 receptor was expressed in nonpigmented epithelial cells of the ciliary body and controlled aqueous humor dynamics. P2Y6KO mice exhibited sustained elevation of IOP, age-dependent damage to the optic nerve, thinning of ganglion cell plus inner plexiform layers, and a reduction of RGC numbers. These changes in P2Y6KO mice were attenuated by an IOP lowering agent. Consistent with RGC damage, visual functions were impaired in middle-aged P2Y6KO mice. We also found that expression and function of P2Y6 receptors in WT mice were significantly reduced by aging, another important risk factor for glaucoma. In summary, our data show that dysfunctional purinergic signaling causes IOP dysregulation, resulting in glaucomatous optic neuropathy.
Collapse
Affiliation(s)
- Youichi Shinozaki
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, and
| | - Kenji Kashiwagi
- Department of Ophthalmology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Kazuhiko Namekata
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Akiko Takeda
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, and
| | - Nobuhiko Ohno
- Division of Neurobiology and Bioinformatics, National Institute for Physiological Sciences, Okazaki, Aichi, Japan
| | - Bernard Robaye
- Institute of Interdisciplinary Research and.,Institute of Biology and Molecular Medicine, Université Libre de Bruxelles, Belgium
| | - Takayuki Harada
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Takeshi Iwata
- Division of Molecular and Cellular Biology, National Institute of Sensory Organs, National Hospital Organization Tokyo Medical Center, Tokyo, Japan
| | - Schuichi Koizumi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, and
| |
Collapse
|
94
|
Du M, Phelps E, Balangue MJ, Dockins A, Moiseyev G, Shin Y, Kane S, Otalora L, Ma JX, Farjo R, Farjo KM. Transgenic Mice Over-Expressing RBP4 Have RBP4-Dependent and Light-Independent Retinal Degeneration. Invest Ophthalmol Vis Sci 2017; 58:4375–4383. [PMID: 28813718 PMCID: PMC5560100 DOI: 10.1167/iovs.17-22107] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Purpose Transgenic mice overexpressing serum retinol-binding protein (RBP4-Tg) develop progressive retinal degeneration, characterized by microglia activation, yet the precise mechanisms underlying retinal degeneration are unclear. Previous studies showed RBP4-Tg mice have normal ocular retinoid levels, suggesting that degeneration is independent of the retinoid visual cycle or light exposure. The present study addresses whether retinal degeneration is light-dependent and RBP4-dependent by testing the effects of dark-rearing and pharmacological lowering of serum RBP4 levels, respectively. Methods RBP4-Tg mice reared on normal mouse chow in normal cyclic light conditions were directly compared to RBP4-Tg mice exposed to chow supplemented with the RBP4-lowering compound A1120 or dark-rearing conditions. Quantitative retinal histological analysis was conducted to assess retinal degeneration, and electroretinography (ERG) and optokinetic tracking (OKT) tests were performed to assess retinal and visual function. Ocular retinoids and bis-retinoid A2E were quantified. Results Dark-rearing RBP4-Tg mice effectively reduced ocular bis-retinoid A2E levels, but had no significant effect on retinal degeneration or dysfunction in RBP4-Tg mice, demonstrating that retinal degeneration is light-independent. A1120 treatment lowered serum RBP4 levels similar to wild-type mice, and prevented structural retinal degeneration. However, A1120 treatment did not prevent retinal dysfunction in RBP4-Tg mice. Moreover, RBP4-Tg mice on A1120 diet had significant worsening of OKT response and loss of cone photoreceptors compared to RBP4-Tg mice on normal chow. This may be related to the very significant reduction in retinyl ester levels in the retina of mice on A1120-supplemented diet. Conclusions Retinal degeneration in RBP4-Tg mice is RBP4-dependent and light-independent.
Collapse
Affiliation(s)
- Mei Du
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Eric Phelps
- EyeCRO LLC, Oklahoma City, Oklahoma, United States
| | | | | | - Gennadiy Moiseyev
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Younghwa Shin
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Shelley Kane
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Laura Otalora
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Jian-Xing Ma
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Rafal Farjo
- EyeCRO LLC, Oklahoma City, Oklahoma, United States
| | - Krysten M Farjo
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| |
Collapse
|
95
|
Bian M, Du X, Wang P, Cui J, Xu J, Gu J, Zhang T, Chen Y. Combination of ginsenoside Rb1 and Rd protects the retina against bright light-induced degeneration. Sci Rep 2017; 7:6015. [PMID: 28729651 PMCID: PMC5519667 DOI: 10.1038/s41598-017-06471-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 06/14/2017] [Indexed: 01/17/2023] Open
Abstract
Photoreceptor degeneration is a central pathology of various retinal degenerative diseases which currently lack effective therapies. Antioxidant and anti-inflammatory activities are noted for Panax notoginsenoside saponins (PNS) and related saponin compound(s). However, the photoreceptor protective potentials of PNS or related saponin compound(s) remain unknown. The current study revealed that PNS protected against photoreceptor loss in bright light-exposed BALB/c mice. Combination of ginsenoside Rb1 and Rd, two major saponin compounds of PNS, recapitulated the retinal protection of PNS and attenuated retinal oxidative stress and inflammatory changes. Rb1 or Rd partially alleviated all-trans-Retinal-induced oxidative stress in ARPE19 cells. Rb1 or Rd suppressed lipopolysaccharides (LPS)-induced proinflammatory gene expression in ARPE19 and RAW264.7 cells. Rb1 or Rd also modulated the expression of proinflammatory microRNA, miR-155 and its direct target, anti-inflammatory SHIP1, in LPS-stimulated RAW264.7 cells. The retinal expression of miR-155 and SHIP1 was altered preceding extensive retinal damage, which was maintained at normal level by Rb1 and Rd combination. This work shows for the first time that altered expression of miR-155 and SHIP1 are involved in photoreceptor degeneration. Most importantly, novel retinal protective activities of combination of Rb1 and Rd justify further evaluation for the treatment of related retinal degenerative disorders.
Collapse
Affiliation(s)
- Minjuan Bian
- Yueyang Hospital and Clinical Research Institute of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Xiaoye Du
- Yueyang Hospital and Clinical Research Institute of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Peiwei Wang
- Yueyang Hospital and Clinical Research Institute of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Jingang Cui
- Yueyang Hospital and Clinical Research Institute of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Jing Xu
- Department of Pharmacy, East China University of Science and Technology, Shanghai, 201203, China
| | - Jiangping Gu
- Department of Pharmacy, East China University of Science and Technology, Shanghai, 201203, China
| | - Teng Zhang
- Yueyang Hospital and Clinical Research Institute of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China.
| | - Yu Chen
- Yueyang Hospital and Clinical Research Institute of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China.
| |
Collapse
|
96
|
Karlstetter M, Dannhausen K, Langmann T. Mikroglia und Immuntherapien bei degenerativen Netzhauterkrankungen. MED GENET-BERLIN 2017. [DOI: 10.1007/s11825-017-0132-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Zusammenfassung
Bei allen bisher im Detail untersuchten erblichen Netzhautdegenerationen liegt eine dem Erkrankungsverlauf abträgliche chronische Aktivierung des angeborenen Immunsystems zugrunde. Vor allem residente Mikrogliazellen der Netzhaut und verschiedene Proteine des löslichen Komplementsystems tragen zu einer Schädigung von Photorezeptoren und retinalem Pigmentepithel bei. Sowohl spezifische Zielstrukturen auf reaktiven Immunzellen als auch fehlregulierte lösliche Immunmodulatoren bieten neue Ansatzpunkte für Therapien, um das Überleben der Netzhaut trotz genetischer Prädisposition zur Degeneration zu fördern. Dieser Beitrag gibt Einblick in die wesentlichen Regulationsmechanismen der Netzhautimmunologie, diskutiert die mögliche Verwendung immunologischer Biomarker für die Netzhautdiagnostik und zeigt immunmodulierende Therapieansätze durch Biologika und endogene Botenstoffe auf.
Collapse
Affiliation(s)
- Marcus Karlstetter
- Aff1 0000 0000 8852 305X grid.411097.a Lehrstuhl für Experimentelle Immunologie des Auges, Zentrum für Augenheilkunde Uniklinik Köln Joseph-Stelzmann-Str. 9 50931 Köln Deutschland
| | - Katharina Dannhausen
- Aff1 0000 0000 8852 305X grid.411097.a Lehrstuhl für Experimentelle Immunologie des Auges, Zentrum für Augenheilkunde Uniklinik Köln Joseph-Stelzmann-Str. 9 50931 Köln Deutschland
| | - Thomas Langmann
- Aff1 0000 0000 8852 305X grid.411097.a Lehrstuhl für Experimentelle Immunologie des Auges, Zentrum für Augenheilkunde Uniklinik Köln Joseph-Stelzmann-Str. 9 50931 Köln Deutschland
| |
Collapse
|
97
|
Parmar T, Parmar VM, Arai E, Sahu B, Perusek L, Maeda A. Acute Stress Responses Are Early Molecular Events of Retinal Degeneration in Abca4-/-Rdh8-/- Mice After Light Exposure. Invest Ophthalmol Vis Sci 2017; 57:3257-67. [PMID: 27315541 PMCID: PMC4928696 DOI: 10.1167/iovs.15-18993] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
PURPOSE Mice lacking ATP-binding cassette transporter 4 (ABCA4) and retinol dehydrogenase 8 (RDH8) mimic features of human Stargardt disease and age-related macular degeneration. RNA-sequencing of whole eyes was done to study early gene expression changes in Abca4-/-Rdh8-/- mice. METHODS Abca4-/-Rdh8-/- mice at 4 weeks of age were exposed to intense light. Total RNA was extracted from whole eyes and used to generate RNA libraries that were paired-end sequenced on the Illumina HiSeq 2500 device. Differentially expressed genes were annotated using Gene set enrichment analysis (GSEA). Selected genes in enriched pathways exhibiting differential expression were validated using quantitative qRT-PCR and ELISA. RESULTS Transcriptome analysis of the whole eye identified 200 genes that were differentially expressed 24 hours after light exposure compared to no light in Abca4-/-Rdh8-/- mice. Expression of several visual cycle and photoreceptor genes were decreased, indicative of photoreceptor/RPE cell death. Gene categories of early stress response genes, inflammatory cytokines, immune factors, and JAK STAT components were upregulated. Lipocalin 2 (Lcn2) was the most upregulated early stress response gene identified. Protein LCN2 was produced by RPE cells and the neural retina after intense light exposure as well as in cultured RPE cells from mice and humans incubated with lipopolysaccharide or photoreceptor outer segments. CONCLUSIONS Identification of important mediators involved in the crosstalk between the acute stress response and immune activation in RPE cells and the neural retina, such as LCN2, provide novel molecular targets for reducing cellular stress during retinal degeneration.
Collapse
Affiliation(s)
- Tanu Parmar
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, Ohio, United States
| | - Vipul M Parmar
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, Ohio, United States
| | - Eisuke Arai
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, Ohio, United States
| | - Bhubanananda Sahu
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, Ohio, United States
| | - Lindsay Perusek
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, Ohio, United States
| | - Akiko Maeda
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, Ohio, United States 2Department of Pharmacology, Case Western Reserve University, Cleveland, Ohio, United States
| |
Collapse
|
98
|
Mendiola AS, Cardona AE. The IL-1β phenomena in neuroinflammatory diseases. J Neural Transm (Vienna) 2017; 125:781-795. [PMID: 28534174 DOI: 10.1007/s00702-017-1732-9] [Citation(s) in RCA: 139] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 05/04/2017] [Indexed: 02/06/2023]
Abstract
It is becoming increasingly clear that neuroinflammation has a causal role in the pathogenesis of central nervous system (CNS)-related diseases, and therefore therapeutic strategies targeting the regulation or availability of inflammatory mediators can be used to prevent or mitigate pathology. Interestingly, the proinflammatory cytokine, interleukin-1 beta (IL-1β), has been implicated in perpetuating immune responses and contributing to disease severity in a variety of CNS diseases ranging from multiple sclerosis, neurodegenerative diseases, traumatic brain injury, and diabetic retinopathy. Moreover, pharmacological blockade of IL-1 signaling has shown to be beneficial in some autoimmune and autoinflammatory diseases, making IL-1β a promising therapeutic target in neuroinflammatory conditions. This review highlights recent advances of our understanding on the multifaceted roles of IL-1β in neuroinflammatory diseases.
Collapse
Affiliation(s)
- Andrew S Mendiola
- Department of Biology, University of Texas at San Antonio, San Antonio, TX, 78249, USA
| | - Astrid E Cardona
- Department of Biology, University of Texas at San Antonio, San Antonio, TX, 78249, USA.
| |
Collapse
|
99
|
Tonade D, Liu H, Kern TS. Photoreceptor Cells Produce Inflammatory Mediators That Contribute to Endothelial Cell Death in Diabetes. Invest Ophthalmol Vis Sci 2017; 57:4264-71. [PMID: 27548900 PMCID: PMC5015981 DOI: 10.1167/iovs.16-19859] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
PURPOSE Recent studies suggest that photoreceptor cells regulate local inflammation in the retina in diabetes. The purpose of this study was to determine if photoreceptor cells themselves produce inflammatory proteins in diabetes and if soluble factors released by photoreceptors in elevated glucose induce inflammatory changes in nearby cells. METHODS Laser capture microdissection was used to isolate the outer retina (photoreceptors) from the inner retina in nondiabetic and diabetic mice. Diabetes-induced changes in the expression of inflammatory targets were assessed by reverse transcription polymerase chain reaction and immunohistochemistry. Cell culture experiments were carried out to determine if photoreceptors in vitro and ex vivo release soluble mediators that can stimulate nearby cells. Photoreceptor contribution to leukocyte-mediated endothelial cell death was tested using coculture models. RESULTS Messenger ribonucleic acid and protein expression levels for inflammatory proteins intercellular adhesion molecule 1 (ICAM1), inducible nitric oxide synthase (iNOS), and cyclooxygenase 2 (COX2) were increased in photoreceptors cells in diabetes. In vitro and ex vivo studies show that photoreceptor cells in elevated glucose release mediators that can induce tumor necrosis factor-α in leukocytes and endothelial cells, but not in glia. The soluble mediators released by photoreceptor cells in elevated glucose are regulated by transforming growth factor β-activated kinase 1 and nicotinamide adenine dinucleotide phosphate oxidase (NADPH oxidase) signaling. In contrast to enhanced leukocyte-mediated killing of endothelial cells by leukocytes from wild-type diabetic mice, leukocytes from diabetic mice lacking photoreceptor cells (opsin-/-) did not kill endothelial cells. CONCLUSIONS These data indicate that photoreceptor cells are a source of inflammatory proteins in diabetes, and their release of soluble mediators can contribute to the death of retinal capillaries in diabetes.
Collapse
Affiliation(s)
- Deoye Tonade
- Department of Pharmacology Case Western Reserve University, Cleveland, Ohio, United States
| | - Haitao Liu
- Department of Medicine, Case Western Reserve University, Cleveland, Ohio, United States
| | - Timothy S Kern
- Department of Pharmacology Case Western Reserve University, Cleveland, Ohio, United States 2Department of Medicine, Case Western Reserve University, Cleveland, Ohio, United States 3Veterans Administration Medical Center Research Service, Cleveland, Ohio, United States
| |
Collapse
|
100
|
Du M, Martin A, Hays F, Johnson J, Farjo RA, Farjo KM. Serum retinol-binding protein-induced endothelial inflammation is mediated through the activation of toll-like receptor 4. Mol Vis 2017; 23:185-197. [PMID: 28400700 PMCID: PMC5373036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 03/29/2017] [Indexed: 11/02/2022] Open
Abstract
PURPOSE Elevation of serum retinol-binding protein 4 (RBP4) induces inflammation in primary human retinal microvascular endothelial cells (HRECs) via a retinol-independent mechanism; thus, it may play a causative role in the development and progression of vascular lesions in diabetic retinopathy (DR). Since HRECs do not express the classical RBP4 receptor, stimulated by retinoic acid gene 6 (STRA6), this study focuses on identifying the endothelial cell receptor and signaling that mediate RBP4-induced inflammation. METHODS HRECs were treated with a toll-like receptor 4 (TLR4) small molecule inhibitor (Cli95, also known as TAK-242), TLR4 neutralizing antibody, or mitogen-activated protein kinase (MAPK) inhibitors before treatment with purified recombinant RBP4. The HREC inflammatory response was quantified by in vitro leukostasis assays, western blotting, and enzyme-linked immunosorbent assay (ELISA). To understand how the serum binding partner for RBP4, transthyretin (TTR), may affect RBP4 activity, we also measured RBP4 and TTR levels in serum and retinal lysates from RBP4-Tg and wild-type mice. RESULTS TLR4 inhibition significantly reduced RBP4-induced expression of pro-inflammatory proteins and in vitro leukostasis. RBP4 treatment significantly increased phosphoactivation of p38 and c-Jun N-terminal protein kinase (JNK). The p38 inhibitor (SB203580) attenuated RBP4-stimulated vascular cell adhesion molecule 1 (VCAM-1), intracellular adhesion molecule 1 (ICAM-1), monocyte chemoattractant protein (MCP-1), and interleukin 6 (IL-6) production, while the JNK inhibitor (SP600125) reduced RBP4-stimulated sICAM-1, endothelial cell selectin (E-selectin), and MCP-1 production. The MAPK inhibitors only showed partial (50-70%) suppression of the RBP4-stimulated proinflammatory response. Moreover, TLR4 inhibition did not decrease RBP4-induced MAPK phosphoactivation, suggesting that RBP4-mediated MAPK activation is TLR4 independent and occurs through a secondary unknown receptor. We also found that the RBP4/TTR molar ratio was exceptionally high in the retina of RBP4-Tg mice, indicating an abundance of TTR-free RBP4. CONCLUSIONS RBP4-induced inflammation is largely mediated by TLR4, and in part, through JNK and p38 MAPK signaling. The high TTR/RBP4 molar ratio in serum likely protects the endothelium from the proinflammatory effects of RBP4 in vivo, whereas elevation of serum RBP4 causes a significant increase in TTR-free RBP4 in retinal tissue. This offers insight into how RBP4-Tg mice can develop retinal neurodegeneration without coincident retinal microvascular pathology.
Collapse
Affiliation(s)
- Mei Du
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK,Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Ashley Martin
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK,Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Franklin Hays
- Department of Biochemistry, University of Oklahoma Health Sciences Center, Oklahoma City, OK,Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Jennifer Johnson
- Department of Biochemistry, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | | | - Krysten M. Farjo
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK,Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| |
Collapse
|