1
|
Ufret-Vincenty RL, Kirman DC, Ulker-Yilmazer G, Aredo B, Shrestha S, Turpin E, Yuksel S, Zegeye Y, Ludwig S, Moresco EMY, He YG, Beutler B. Nonredundant Role of Leishmanolysin-Like (Lmln) Zinc-Metallopeptidase in Retinal Homeostasis. Am J Ophthalmol 2025; 269:147-160. [PMID: 39209209 DOI: 10.1016/j.ajo.2024.08.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 08/15/2024] [Accepted: 08/15/2024] [Indexed: 09/04/2024]
Abstract
PURPOSE To determine if Lmln, a Zinc-metallopeptidase, is important for retinal homeostasis. DESIGN Basic research in mouse models of retinal degeneration. METHODS Combining an unbiased N-ethyl-N-nitrosourea mutagenesis pipeline in mice with optical coherence tomography (OCT) screening and automated meiotic mapping, we identified an allele (nemeth) that seemed to be associated with outer nuclear layer (ONL) thinning. Since nemeth was predicted to lead to a nonsense mutation of the Lmln gene, we targeted Lmln using CRISPR/Cas-9 technology and characterized the impact on retinal anatomy and function. RESULTS OCT imaging demonstrated an outer retinal degeneration in Lmln-/- mice (P = 7.3 × 10-9 for ONL at 2 m) that progressed over the first 6 months of life and then stabilized. Light microscopy showed loss of ONL nuclei (P ranged between .00033 and .0097 for posterior measurements), and a TUNEL assay revealed a small but significant increase in apoptosis (P = .034). Lmln-/- mice accumulated fundus spots (P = .0030 by 2 m of age) and activated subretinal microglia (P ranged from .0007 to 8 × 10-13 for Gal3+ cells). Scotopic electroretinography demonstrated a decrease in retinal function in Lmln-/- mice both at 6 m (only a-wave, P < .01 for all stimuli) and at 10 m of age (P < .01 for both a-wave and b-wave with all stimuli). CONCLUSIONS Our work revealed a previously unknown essential role for Lmln in maintaining retinal anatomy and function. Further studies using this new model will be aimed at determining the cellular expression of Lmln and its mechanisms of action within the retina.
Collapse
Affiliation(s)
- Rafael L Ufret-Vincenty
- Department of Ophthalmology (R.L.U.V., D.C.K., G.U.Y., B.A., S.S., E.T., S.Y., Y.Z., Y.G.H.), UT Southwestern Medical Center, Dallas, Texas, USA.
| | - Dogan Can Kirman
- Department of Ophthalmology (R.L.U.V., D.C.K., G.U.Y., B.A., S.S., E.T., S.Y., Y.Z., Y.G.H.), UT Southwestern Medical Center, Dallas, Texas, USA
| | - Gizem Ulker-Yilmazer
- Department of Ophthalmology (R.L.U.V., D.C.K., G.U.Y., B.A., S.S., E.T., S.Y., Y.Z., Y.G.H.), UT Southwestern Medical Center, Dallas, Texas, USA
| | - Bogale Aredo
- Department of Ophthalmology (R.L.U.V., D.C.K., G.U.Y., B.A., S.S., E.T., S.Y., Y.Z., Y.G.H.), UT Southwestern Medical Center, Dallas, Texas, USA
| | - Sangita Shrestha
- Department of Ophthalmology (R.L.U.V., D.C.K., G.U.Y., B.A., S.S., E.T., S.Y., Y.Z., Y.G.H.), UT Southwestern Medical Center, Dallas, Texas, USA
| | - Emily Turpin
- Department of Ophthalmology (R.L.U.V., D.C.K., G.U.Y., B.A., S.S., E.T., S.Y., Y.Z., Y.G.H.), UT Southwestern Medical Center, Dallas, Texas, USA
| | - Seher Yuksel
- Department of Ophthalmology (R.L.U.V., D.C.K., G.U.Y., B.A., S.S., E.T., S.Y., Y.Z., Y.G.H.), UT Southwestern Medical Center, Dallas, Texas, USA
| | - Yeshumenesh Zegeye
- Department of Ophthalmology (R.L.U.V., D.C.K., G.U.Y., B.A., S.S., E.T., S.Y., Y.Z., Y.G.H.), UT Southwestern Medical Center, Dallas, Texas, USA
| | - Sara Ludwig
- Center for the Genetics of Host Defense (S.L., E.M.Y.M., B.B.), UT Southwestern Medical Center, Dallas, Texas, USA
| | - Eva Marie Y Moresco
- Center for the Genetics of Host Defense (S.L., E.M.Y.M., B.B.), UT Southwestern Medical Center, Dallas, Texas, USA
| | - Yu-Guang He
- Department of Ophthalmology (R.L.U.V., D.C.K., G.U.Y., B.A., S.S., E.T., S.Y., Y.Z., Y.G.H.), UT Southwestern Medical Center, Dallas, Texas, USA
| | - Bruce Beutler
- Center for the Genetics of Host Defense (S.L., E.M.Y.M., B.B.), UT Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
2
|
Ji J, Xiong C, Yang H, Jiang Z, Zhang Y, Wang X, Yu T, Li Q, Zhu S, Zhou Y. The aryl hydrocarbon receptor: A crucial mediator in ocular disease pathogenesis and therapeutic target. Exp Eye Res 2024; 249:110144. [PMID: 39486499 DOI: 10.1016/j.exer.2024.110144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 10/07/2024] [Accepted: 10/29/2024] [Indexed: 11/04/2024]
Abstract
The aryl hydrocarbon receptor (AHR) is a pivotal nuclear receptor involved in mediating cellular responses to a wide range of environmental pollutants and endogenous ligands. AHR plays a central role in regulating essential physiological processes, including xenobiotic metabolism, immune response modulation, cell cycle control, tumorigenesis, and developmental events. Recent studies have identified AHR as a critical mediator and a potential therapeutic target in the pathogenesis of ocular diseases. This review provides a thorough analysis of the various functions of AHR signalling in the ocular environment, with a specific emphasis on its effects on the retina, retinal pigment epithelium (RPE), choroid, and cornea. We provide a detailed discussion on the molecular mechanisms through which AHR integrates environmental and endogenous signals, influencing the development and progression of age-related macular degeneration (AMD), retinitis pigmentosa, uveitis, and other major ocular disorders. Furthermore, we evaluate the therapeutic potential of modulating AHR activity through novel ligands and agonists as a strategy for treating eye diseases. Understanding the molecular mechanisms of AHR in ocular tissues may facilitate the development of AHR-targeted therapies, which is crucial for addressing the pressing clinical demand for novel treatment strategies in ocular diseases.
Collapse
Affiliation(s)
- Juanjuan Ji
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Genome Sequencing Center, Department of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Chanyu Xiong
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Genome Sequencing Center, Department of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Huining Yang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Genome Sequencing Center, Department of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Zhilin Jiang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Genome Sequencing Center, Department of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yun Zhang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Genome Sequencing Center, Department of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiao Wang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Genome Sequencing Center, Department of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Tianshu Yu
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Genome Sequencing Center, Department of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Qiong Li
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Genome Sequencing Center, Department of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Shikai Zhu
- Organ Transplant Center, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yu Zhou
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Genome Sequencing Center, Department of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
3
|
Banna HU, Slayo M, Armitage JA, Del Rosal B, Vocale L, Spencer SJ. Imaging the eye as a window to brain health: frontier approaches and future directions. J Neuroinflammation 2024; 21:309. [PMID: 39614308 PMCID: PMC11606158 DOI: 10.1186/s12974-024-03304-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 11/18/2024] [Indexed: 12/01/2024] Open
Abstract
Recent years have seen significant advances in diagnostic testing of central nervous system (CNS) function and disease. However, there remain challenges in developing a comprehensive suite of non- or minimally invasive assays of neural health and disease progression. Due to the direct connection with the CNS, structural changes in the neural retina, retinal vasculature and morphological changes in retinal immune cells can occur in parallel with disease conditions in the brain. The retina can also, uniquely, be assessed directly and non-invasively. For these reasons, the retina may prove to be an important "window" for revealing and understanding brain disease. In this review, we discuss the gross anatomy of the eye, focusing on the sensory and non-sensory cells of the retina, especially microglia, that lend themselves to diagnosing brain disease by imaging the retina. We include a history of ocular imaging to describe the different imaging approaches undertaken in the past and outline current and emerging technologies including retinal autofluorescence imaging, Raman spectroscopy, and artificial intelligence image analysis. These new technologies show promising potential for retinal imaging to be used as a tool for the diagnosis of brain disorders such as Alzheimer's disease and others and the assessment of treatment success.
Collapse
Affiliation(s)
- Hasan U Banna
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Melbourne, VIC, Australia
| | - Mary Slayo
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Melbourne, VIC, Australia
- Institute of Veterinary Physiology and Biochemistry, Justus Liebig University, Giessen, Germany
| | - James A Armitage
- School of Medicine (Optometry), Deakin University, Waurn Ponds, VIC, Australia
| | | | - Loretta Vocale
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Melbourne, VIC, Australia
| | - Sarah J Spencer
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Melbourne, VIC, Australia.
| |
Collapse
|
4
|
Jiang H, Yang J, Fu Q, Li A, Qin H, Liu M. Induction of Endoplasmic Reticulum Stress and Aryl Hydrocarbon Receptor Pathway Expression by Blue LED Irradiation in Oral Squamous Cell Carcinoma. JOURNAL OF BIOPHOTONICS 2024; 17:e202400226. [PMID: 39209312 DOI: 10.1002/jbio.202400226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 07/03/2024] [Accepted: 07/27/2024] [Indexed: 09/04/2024]
Abstract
Photobiomodulation therapy, as an emerging treatment modality, has been widely used in dentistry. However, reports on blue light therapy for oral cancer are scarce. This study investigated the effects of 457 and 475 nm LED irradiation on SCC-25 cells and explored the potential mechanisms underlying the impact of blue light. Both wavelengths were found to inhibit cell viability, induce oxidative stress, and cause cell cycle arrest without leading to cell death. Notably, the inhibitory effect of 457 nm blue light on cell proliferation was more sustained. Transcriptome sequencing was performed to explore the underlying mechanisms, revealing that blue light induced endoplasmic reticulum stress in SCC-25 cells, with 457 nm light showing a more pronounced effect. Moreover, 457 nm blue light upregulated the expression of the aryl hydrocarbon receptor pathway, indicating potential therapeutic prospects for the combined use of blue light and pharmacological agents.
Collapse
Affiliation(s)
- Hui Jiang
- Academy for Engineering and Technology, Fudan University, Shanghai, China
| | - Jiali Yang
- School of Information Science and Technology, Fudan University, Shanghai, China
| | - Qiqi Fu
- School of Information Science and Technology, Fudan University, Shanghai, China
| | - Angze Li
- School of Information Science and Technology, Fudan University, Shanghai, China
| | - Haokuan Qin
- Academy for Engineering and Technology, Fudan University, Shanghai, China
| | - Muqing Liu
- School of Information Science and Technology, Fudan University, Shanghai, China
- Zhongshan DB-Light Technology Co., Ltd, Zhongshan City, Guangdong Province, China
| |
Collapse
|
5
|
Xie L, Wu Q, Li K, Khan MAS, Zhang A, Sinha B, Li S, Chang SL, Brody DL, Grinstaff MW, Zhou S, Alterovitz G, Liu P, Wang X. Tryptophan Metabolism in Alzheimer's Disease with the Involvement of Microglia and Astrocyte Crosstalk and Gut-Brain Axis. Aging Dis 2024; 15:2168-2190. [PMID: 38916729 PMCID: PMC11346405 DOI: 10.14336/ad.2024.0134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 05/03/2024] [Indexed: 06/26/2024] Open
Abstract
Alzheimer's disease (AD) is an age-dependent neurodegenerative disease characterized by extracellular Amyloid Aβ peptide (Aβ) deposition and intracellular Tau protein aggregation. Glia, especially microglia and astrocytes are core participants during the progression of AD and these cells are the mediators of Aβ clearance and degradation. The microbiota-gut-brain axis (MGBA) is a complex interactive network between the gut and brain involved in neurodegeneration. MGBA affects the function of glia in the central nervous system (CNS), and microbial metabolites regulate the communication between astrocytes and microglia; however, whether such communication is part of AD pathophysiology remains unknown. One of the potential links in bilateral gut-brain communication is tryptophan (Trp) metabolism. The microbiota-originated Trp and its metabolites enter the CNS to control microglial activation, and the activated microglia subsequently affect astrocyte functions. The present review highlights the role of MGBA in AD pathology, especially the roles of Trp per se and its metabolism as a part of the gut microbiota and brain communications. We (i) discuss the roles of Trp derivatives in microglia-astrocyte crosstalk from a bioinformatics perspective, (ii) describe the role of glia polarization in the microglia-astrocyte crosstalk and AD pathology, and (iii) summarize the potential of Trp metabolism as a therapeutic target. Finally, we review the role of Trp in AD from the perspective of the gut-brain axis and microglia, as well as astrocyte crosstalk, to inspire the discovery of novel AD therapeutics.
Collapse
Affiliation(s)
- Lushuang Xie
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA.
- Acupuncture and Moxibustion College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610075, China.
| | - Qiaofeng Wu
- Acupuncture and Moxibustion College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610075, China.
| | - Kelin Li
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA.
- Department of Chemistry, Boston University, Boston, MA 02215, USA.
| | - Mohammed A. S. Khan
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - Andrew Zhang
- Biomedical Cybernetics Laboratory, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - Bharati Sinha
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - Sihui Li
- Acupuncture and Moxibustion College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610075, China.
| | - Sulie L. Chang
- Department of Biological Sciences, Institute of NeuroImmune Pharmacology, Seton Hall University, South Orange, NJ 07079, USA.
| | - David L. Brody
- Department of Neurology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA.
| | | | - Shuanhu Zhou
- Harvard Medical School, Harvard Stem Cell Institute, Boston, MA 02115, USA.
| | - Gil Alterovitz
- Biomedical Cybernetics Laboratory, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - Pinghua Liu
- Department of Chemistry, Boston University, Boston, MA 02215, USA.
| | - Xin Wang
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
6
|
Guarnieri T. Light Sensing beyond Vision: Focusing on a Possible Role for the FICZ/AhR Complex in Skin Optotransduction. Cells 2024; 13:1082. [PMID: 38994936 PMCID: PMC11240502 DOI: 10.3390/cells13131082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/16/2024] [Accepted: 06/20/2024] [Indexed: 07/13/2024] Open
Abstract
Although our skin is not the primary visual organ in humans, it acts as a light sensor, playing a significant role in maintaining our health and overall well-being. Thanks to the presence of a complex and sophisticated optotransduction system, the skin interacts with the visible part of the electromagnetic spectrum and with ultraviolet (UV) radiation. Following a brief overview describing the main photosensitive molecules that detect specific electromagnetic radiation and their associated cell pathways, we analyze their impact on physiological functions such as melanogenesis, immune response, circadian rhythms, and mood regulation. In this paper, we focus on 6-formylindolo[3,2-b]carbazole (FICZ), a photo oxidation derivative of the essential amino acid tryptophan (Trp). This molecule is the best endogenous agonist of the Aryl hydrocarbon Receptor (AhR), an evolutionarily conserved transcription factor, traditionally recognized as a signal transducer of both exogenous and endogenous chemical signals. Increasing evidence indicates that AhR is also involved in light sensing within the skin, primarily due to its ligand FICZ, which acts as both a chromophore and a photosensitizer. The biochemical reactions triggered by their interaction impact diverse functions and convey crucial data to our body, thus adding a piece to the complex puzzle of pathways that allow us to decode and elaborate environmental stimuli.
Collapse
Affiliation(s)
- Tiziana Guarnieri
- Cell Physiology Laboratory, Department of Biological, Geological, and Environmental Sciences, Alma Mater Studiorum Università di Bologna, Via Francesco Selmi 3, 40126 Bologna, Italy;
- Interdepartmental Center for Industrial Research in Life Sciences and Technologies, University of Bologna, 40126 Bologna, Italy
- Consiglio Nazionale delle Ricerche, Istituto per le Applicazioni del Calcolo “Mauro Picone”, Via dei Taurini 19, 00185 Roma, Italy
| |
Collapse
|
7
|
Schustak J, Han H, Bond K, Huang Q, Saint-Geniez M, Bao Y. Phenotypic high-throughput screening identifies aryl hydrocarbon receptor agonism as common inhibitor of toxin-induced retinal pigment epithelium cell death. PLoS One 2024; 19:e0301239. [PMID: 38635505 PMCID: PMC11025755 DOI: 10.1371/journal.pone.0301239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 03/12/2024] [Indexed: 04/20/2024] Open
Abstract
The retinal pigment epithelium (RPE) is essential to maintain retinal function, and RPE cell death represents a key pathogenic stage in the progression of several blinding ocular diseases, including age-related macular degeneration (AMD). To identify pathways and compounds able to prevent RPE cell death, we developed a phenotypic screening pipeline utilizing a compound library and high-throughput screening compatible assays on the human RPE cell line, ARPE-19, in response to different disease relevant cytotoxic stimuli. We show that the metabolic by-product of the visual cycle all-trans-retinal (atRAL) induces RPE apoptosis, while the lipid peroxidation by-product 4-hydroxynonenal (4-HNE) promotes necrotic cell death. Using these distinct stimuli for screening, we identified agonists of the aryl hydrocarbon receptor (AhR) as a consensus target able to prevent both atRAL mediated apoptosis and 4-HNE-induced necrotic cell death. This works serves as a framework for future studies dedicated to screening for inhibitors of cell death, as well as support for the discussion of AhR agonism in RPE pathology.
Collapse
Affiliation(s)
- Joshua Schustak
- Department of Ophthalmology, BioMedical Research, Novartis, Cambridge, Massachusetts, United States of America
| | - Hongwei Han
- Department of Ophthalmology, BioMedical Research, Novartis, Cambridge, Massachusetts, United States of America
| | - Kyle Bond
- Department of Ophthalmology, BioMedical Research, Novartis, Cambridge, Massachusetts, United States of America
| | - Qian Huang
- Department of Ophthalmology, BioMedical Research, Novartis, Cambridge, Massachusetts, United States of America
| | - Magali Saint-Geniez
- Department of Ophthalmology, BioMedical Research, Novartis, Cambridge, Massachusetts, United States of America
| | - Yi Bao
- Department of Ophthalmology, BioMedical Research, Novartis, Cambridge, Massachusetts, United States of America
| |
Collapse
|
8
|
Zegeye Y, Aredo B, Yuksel S, Kirman DC, Kumar A, Chen B, Turpin E, Shresta S, He YG, Gautron L, Tang M, Li X, DiCesare SM, Hulleman JD, Xing C, Ludwig S, Moresco EMY, Beutler BA, Ufret-Vincenty RL. E3 ubiquitin ligase Herc3 deficiency leads to accumulation of subretinal microglia and retinal neurodegeneration. Sci Rep 2024; 14:3010. [PMID: 38321224 PMCID: PMC10847449 DOI: 10.1038/s41598-024-53731-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 02/04/2024] [Indexed: 02/08/2024] Open
Abstract
Activated microglia have been implicated in the pathogenesis of age-related macular degeneration (AMD), diabetic retinopathy, and other neurodegenerative and neuroinflammatory disorders, but our understanding of the mechanisms behind their activation is in infant stages. With the goal of identifying novel genes associated with microglial activation in the retina, we applied a semiquantitative fundus spot scoring scale to an unbiased, state-of-the-science mouse forward genetics pipeline. A mutation in the gene encoding the E3 ubiquitin ligase Herc3 led to prominent accumulation of fundus spots. CRISPR mutagenesis was used to generate Herc3-/- mice, which developed prominent accumulation of fundus spots and corresponding activated Iba1 + /CD16 + subretinal microglia, retinal thinning on OCT and histology, and functional deficits by Optomotory and electrophysiology. Bulk RNA sequencing identified activation of inflammatory pathways and differentially expressed genes involved in the modulation of microglial activation. Thus, despite the known expression of multiple E3 ubiquitin ligases in the retina, we identified a non-redundant role for Herc3 in retinal homeostasis. Our findings are significant given that a dysregulated ubiquitin-proteasome system (UPS) is important in prevalent retinal diseases, in which activated microglia appear to play a role. This association between Herc3 deficiency, retinal microglial activation and retinal degeneration merits further study.
Collapse
Affiliation(s)
- Yeshumenesh Zegeye
- Department of Ophthalmology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Bogale Aredo
- Department of Ophthalmology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Seher Yuksel
- Department of Ophthalmology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Dogan Can Kirman
- Department of Ophthalmology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Ashwani Kumar
- McDermott Center for Human Growth and Development, UT Southwestern Medical Center, Dallas, TX, USA
| | - Bo Chen
- Department of Ophthalmology, UT Southwestern Medical Center, Dallas, TX, USA
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Emily Turpin
- Department of Ophthalmology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Sangita Shresta
- Department of Ophthalmology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Yu-Guang He
- Department of Ophthalmology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Laurent Gautron
- Center for Hypothalamic Research and Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Miao Tang
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Xiaohong Li
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Sophia M DiCesare
- Department of Ophthalmology, UT Southwestern Medical Center, Dallas, TX, USA
| | - John D Hulleman
- Department of Ophthalmology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Chao Xing
- McDermott Center for Human Growth and Development, UT Southwestern Medical Center, Dallas, TX, USA
- Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX, USA
| | - Sara Ludwig
- Center for Hypothalamic Research and Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Eva Marie Y Moresco
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Bruce A Beutler
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | | |
Collapse
|
9
|
Yuksel S, Aredo B, Zegeye Y, Zhao CX, Tang M, Li X, Hulleman JD, Gautron L, Ludwig S, Moresco EMY, Butovich IA, Beutler BA, Ufret-Vincenty RL. Forward genetic screening using fundus spot scale identifies an essential role for Lipe in murine retinal homeostasis. Commun Biol 2023; 6:533. [PMID: 37198396 PMCID: PMC10192353 DOI: 10.1038/s42003-023-04870-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 04/24/2023] [Indexed: 05/19/2023] Open
Abstract
Microglia play a role in the pathogenesis of many retinal diseases. Fundus spots in mice often correlate with the accumulation of activated subretinal microglia. Here we use a semiquantitative fundus spot scoring scale in combination with an unbiased, state-of-the-science forward genetics pipeline to identify causative associations between chemically induced mutations and fundus spot phenotypes. Among several associations, we focus on a missense mutation in Lipe linked to an increase in yellow fundus spots in C57BL/6J mice. Lipe-/- mice generated using CRISPR-Cas9 technology are found to develop accumulation of subretinal microglia, a retinal degeneration with decreased visual function, and an abnormal retinal lipid profile. We establish an indispensable role of Lipe in retinal/RPE lipid homeostasis and retinal health. Further studies using this new model will be aimed at determining how lipid dysregulation results in the activation of subretinal microglia and whether these microglia also play a role in the subsequent retinal degeneration.
Collapse
Affiliation(s)
- Seher Yuksel
- Department of Ophthalmology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Bogale Aredo
- Department of Ophthalmology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Yeshumenesh Zegeye
- Department of Ophthalmology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Cynthia X Zhao
- Department of Ophthalmology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Miao Tang
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Xiaohong Li
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - John D Hulleman
- Department of Ophthalmology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Laurent Gautron
- Center for Hypothalamic Research and Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Sara Ludwig
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Eva M Y Moresco
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Igor A Butovich
- Department of Ophthalmology, UT Southwestern Medical Center, Dallas, TX, USA.
| | - Bruce A Beutler
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | | |
Collapse
|
10
|
Choudhary M, Malek G. Potential therapeutic targets for age-related macular degeneration: The nuclear option. Prog Retin Eye Res 2023; 94:101130. [PMID: 36220751 PMCID: PMC10082136 DOI: 10.1016/j.preteyeres.2022.101130] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 09/18/2022] [Accepted: 09/18/2022] [Indexed: 02/07/2023]
Abstract
The functions and activities of nuclear receptors, the largest family of transcription factors in the human genome, have classically focused on their ability to act as steroid and hormone sensors in endocrine organs. However, they are responsible for a diverse array of physiological functions, including cellular homeostasis and metabolism, during development and aging. Though the eye is not a traditional endocrine organ, recent studies have revealed high expression levels of nuclear receptors in cells throughout the posterior pole. These findings have precipitated an interest in investigating the role of these transcription factors in the eye as a function of age and ocular disease, in particular age-related macular degeneration (AMD). As the leading cause of vision impairment in the elderly, identifying signaling pathways that may be targeted for AMD therapy is of great importance, given the lack of therapeutic options for over 85% of patients with this disease. Herein we review this relatively new field and recent findings supporting the hypothesis that the eye is a secondary endocrine organ, in which nuclear receptors serve as the bedrock for biological processes in cells vulnerable in AMD, including retinal pigment epithelial and choroidal endothelial cells, and discuss the therapeutic potential of targeting these receptors for AMD.
Collapse
Affiliation(s)
- Mayur Choudhary
- Duke Eye Center, Department of Ophthalmology, Duke University School of Medicine, Durham, NC, USA
| | - Goldis Malek
- Duke Eye Center, Department of Ophthalmology, Duke University School of Medicine, Durham, NC, USA; Department of Pathology, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
11
|
Naggert ASEN, Collin GB, Wang J, Krebs MP, Chang B. A mouse model of cone photoreceptor function loss (cpfl9) with degeneration due to a mutation in Gucy2e. Front Mol Neurosci 2023; 15:1080136. [PMID: 36698779 PMCID: PMC9868315 DOI: 10.3389/fnmol.2022.1080136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 12/15/2022] [Indexed: 01/11/2023] Open
Abstract
During routine screening of mouse strains and stocks by the Eye Mutant Resource at The Jackson Laboratory for genetic mouse models of human ocular disorders, we identified cpfl9, a mouse model with cone photoreceptor function loss. The mice exhibited an early-onset phenotype that was easily recognized by the absence of a cone-mediated b-wave electroretinography response and by a reduction in rod-mediated photoresponses at four weeks of age. By genetic mapping and high-throughput sequencing of a whole exome capture library of cpfl9, a homozygous 25 bp deletion within exon 11 of the Gucy2e gene was identified, which is predicted to result in a frame shift leading to premature termination. The corresponding protein in human, retinal guanylate cyclase 1 (GUCY2D), plays an important role in rod and cone photoreceptor cell function. Loss-of-function mutations in human GUCY2D cause LCA1, one of the most common forms of Leber congenital amaurosis, which results in blindness at birth or in early childhood. The early loss of cone and reduced rod photoreceptor cell function in the cpfl9 mutant is accompanied by a later, progressive loss of cone and rod photoreceptor cells, which may be relevant to understanding disease pathology in a subset of LCA1 patients and in individuals with cone-rod dystrophy caused by recessive GUCY2D variants. cpfl9 mice will be useful for studying the role of Gucy2e in the retina.
Collapse
|
12
|
Gao S, Zeng Y, Li Y, Cohen ED, Berkowitz BA, Qian H. Fast and slow light-induced changes in murine outer retina optical coherence tomography: complementary high spatial resolution functional biomarkers. PNAS NEXUS 2022; 1:pgac208. [PMID: 36338188 PMCID: PMC9615127 DOI: 10.1093/pnasnexus/pgac208] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 09/21/2022] [Indexed: 11/18/2022]
Abstract
Fast (seconds) and slow (minutes to hours) optical coherence tomography (OCT) responses to light stimulation have been developed to probe outer retinal function with higher spatial resolution than the classical full-field electroretinogram (ERG). However, the relationships between functional information revealed by OCT and ERG are largely unexplored. In this study, we directly compared the fast and slow OCT responses with the ERG. Fast responses [i.e. the optoretinogram (ORG)] are dominated by reflectance changes in the outer segment (OS) and the inner segment ellipsoid zone (ISez). The ORG OS response has faster kinetics and a higher light sensitivity than the ISez response, and both differ significantly with ERG parameters. Sildenafil-inhibition of phototransduction reduced the ORG light sensitivity, suggesting a complete phototransduction pathway is needed for ORG responses. Slower OCT responses were dominated by light-induced changes in the external limiting membrane to retinal pigment epithelium (ELM-RPE) thickness and photoreceptor-tip hyporeflective band (HB) magnitudes, with the biggest changes occurring after prolonged light stimulation. Mice with high (129S6/ev) vs. low (C57BL/6 J) ATP(adenosine triphosphate) synthesis efficiency show similar fast ORG, but dissimilar slow OCT responses. We propose that the ORG reflects passive physiology, such as water movement from photoreceptors, in response to the photocurrent response (measurable by ERG), whereas the slow OCT responses measure mitochondria-driven physiology in the outer retina, such as dark-provoked water removal from the subretinal space.
Collapse
Affiliation(s)
- Shasha Gao
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450001, China
- Visual Function Core, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yong Zeng
- Visual Function Core, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yichao Li
- Visual Function Core, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ethan D Cohen
- Division of Biomedical Physics, Office of Science and Engineering Labs, Center for Devices and Radiological Health, Food and Drug Administration, Silver Spring, MD 20993-0002, USA
| | - Bruce A Berkowitz
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI 48202, USA
| | - Haohua Qian
- Visual Function Core, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
13
|
Grishanova AY, Perepechaeva ML. Aryl Hydrocarbon Receptor in Oxidative Stress as a Double Agent and Its Biological and Therapeutic Significance. Int J Mol Sci 2022; 23:6719. [PMID: 35743162 PMCID: PMC9224361 DOI: 10.3390/ijms23126719] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/14/2022] [Accepted: 06/14/2022] [Indexed: 12/02/2022] Open
Abstract
The aryl hydrocarbon receptor (AhR) has long been implicated in the induction of a battery of genes involved in the metabolism of xenobiotics and endogenous compounds. AhR is a ligand-activated transcription factor necessary for the launch of transcriptional responses important in health and disease. In past decades, evidence has accumulated that AhR is associated with the cellular response to oxidative stress, and this property of AhR must be taken into account during investigations into a mechanism of action of xenobiotics that is able to activate AhR or that is susceptible to metabolic activation by enzymes encoded by the genes that are under the control of AhR. In this review, we examine various mechanisms by which AhR takes part in the oxidative-stress response, including antioxidant and prooxidant enzymes and cytochrome P450. We also show that AhR, as a participant in the redox balance and as a modulator of redox signals, is being increasingly studied as a target for a new class of therapeutic compounds and as an explanation for the pathogenesis of some disorders.
Collapse
Affiliation(s)
| | - Maria L. Perepechaeva
- Federal Research Center of Fundamental and Translational Medicine, Institute of Molecular Biology and Biophysics, Timakova Str. 2, 630117 Novosibirsk, Russia;
| |
Collapse
|
14
|
Tebbe L, Sakthivel H, Makia MS, Kakakhel M, Conley SM, Al-Ubaidi MR, Naash MI. Prph2 disease mutations lead to structural and functional defects in the RPE. FASEB J 2022; 36:e22284. [PMID: 35344225 PMCID: PMC10599796 DOI: 10.1096/fj.202101562rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 02/28/2022] [Accepted: 03/17/2022] [Indexed: 11/11/2022]
Abstract
Prph2 is a photoreceptor-specific tetraspanin with an essential role in the structure and function of photoreceptor outer segments. PRPH2 mutations cause a multitude of retinal diseases characterized by the degeneration of photoreceptors as well as defects in neighboring tissues such as the RPE. While extensive research has analyzed photoreceptors, less attention has been paid to these secondary defects. Here, we use different Prph2 disease models to evaluate the damage of the RPE arising from photoreceptor defects. In Prph2 disease models, the RPE exhibits structural abnormalities and cell loss. Furthermore, RPE functional defects are observed, including impaired clearance of phagocytosed outer segment material and increased microglia activation. The severity of RPE damage is different between models, suggesting that the different abnormal outer segment structures caused by Prph2 disease mutations lead to varying degrees of RPE stress and thus influence the clinical phenotype observed in patients.
Collapse
Affiliation(s)
- Lars Tebbe
- Department of Biomedical Engineering, University of Houston, Houston, Texas, USA
| | - Haarthi Sakthivel
- Department of Biomedical Engineering, University of Houston, Houston, Texas, USA
| | - Mustafa S. Makia
- Department of Biomedical Engineering, University of Houston, Houston, Texas, USA
| | - Mashal Kakakhel
- Department of Biomedical Engineering, University of Houston, Houston, Texas, USA
| | - Shannon M. Conley
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Muayyad R. Al-Ubaidi
- Department of Biomedical Engineering, University of Houston, Houston, Texas, USA
| | - Muna I. Naash
- Department of Biomedical Engineering, University of Houston, Houston, Texas, USA
| |
Collapse
|
15
|
Kim SY, Qian H. Comparison between sodium iodate and lipid peroxide murine models of age-related macular degeneration for drug evaluation-a narrative review. ANNALS OF EYE SCIENCE 2022; 7:8. [PMID: 37622161 PMCID: PMC10448775 DOI: 10.21037/aes-21-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/26/2023]
Abstract
Objective In this review, non-transgenic models of age-related macular degeneration (AMD) are discussed, with focuses on murine retinal degeneration induced by sodium iodate and lipid peroxide (HpODE) as preclinical study platforms. Background AMD is the most common cause of vision loss in a world with an increasingly aging population. The major phenotypes of early and intermediate AMD are increased drusen and autofluorescence, Müller glia activation, infiltrated subretinal microglia and inward moving retinal pigment epithelium cells. Intermediate AMD may progress to advanced AMD, characterized by geography atrophy and/or choroidal neovascularization. Various transgenic and non-transgenic animal models related to retinal degeneration have been generated to investigate AMD pathogenesis and pathobiology, and have been widely used as potential therapeutic evaluation platforms. Methods Two retinal degeneration murine models induced by sodium iodate and HpODE are described. Distinct pathological features and procedures of these two models are compared. In addition, practical protocol and material preparation and assessment methods are elaborated. Conclusion Retina degeneration induced by sodium iodate and HpODE in mouse eye resembles many clinical aspects of human AMD and complimentary to the existent other animal models. However, standardization of procedure and assessment protocols is needed for preclinical studies. Further studies of HpODE on different routes, doses and species will be valuable for the future extensive use. Despite many merits of murine studies, differences between murine and human should be always considered.
Collapse
Affiliation(s)
- Soo-Young Kim
- Department of Pharmaceutics, Department of Biology, Virginia Commonwealth University, Richmond, VA, 23298, USA
- Center for Nanomedicine, Wilmer Eye Institute, Department of Ophthalmology, Johns Hopkins University of Medicine, Baltimore, MD, 21287, USA
| | - Haohua Qian
- Visual Function Core, National Eye Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
16
|
Hammond CL, Roztocil E, Gupta V, Feldon SE, Woeller CF. More than Meets the Eye: The Aryl Hydrocarbon Receptor is an Environmental Sensor, Physiological Regulator and a Therapeutic Target in Ocular Disease. FRONTIERS IN TOXICOLOGY 2022; 4:791082. [PMID: 35295218 PMCID: PMC8915869 DOI: 10.3389/ftox.2022.791082] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 02/08/2022] [Indexed: 12/22/2022] Open
Abstract
The aryl hydrocarbon receptor (AHR) is a ligand activated transcription factor originally identified as an environmental sensor of xenobiotic chemicals. However, studies have revealed that the AHR regulates crucial aspects of cell growth and metabolism, development and the immune system. The importance of the AHR and AHR signaling in eye development, toxicology and disease is now being uncovered. The AHR is expressed in many ocular tissues including the retina, choroid, cornea and the orbit. A significant role for the AHR in age-related macular degeneration (AMD), autoimmune uveitis, and other ocular diseases has been identified. Ligands for the AHR are structurally diverse organic molecules from exogenous and endogenous sources. Natural AHR ligands include metabolites of tryptophan and byproducts of the microbiome. Xenobiotic AHR ligands include persistent environmental pollutants such as dioxins, benzo (a) pyrene [B (a) P] and polychlorinated biphenyls (PCBs). Pharmaceutical agents including the proton pump inhibitors, esomeprazole and lansoprazole, and the immunosuppressive drug, leflunomide, activate the AHR. In this review, we highlight the role of the AHR in the eye and discuss how AHR signaling is involved in responding to endogenous and environmental stimuli. We also present the emerging concept that the AHR is a promising therapeutic target for eye disease.
Collapse
Affiliation(s)
| | | | | | | | - Collynn F. Woeller
- Flaum Eye Institute, Rochester, NY, United States
- Department of Environmental Medicine, School of Medicine and Dentistry, University of Rochester, Rochester, NY, United States
- *Correspondence: Collynn F. Woeller,
| |
Collapse
|
17
|
AlMoallem B, Alharthi E. Novel biallelic AHR splice site mutation cause isolated foveal hypoplasia in Saudi patient: a case report. Ophthalmic Genet 2022; 43:425-429. [DOI: 10.1080/13816810.2022.2039718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Basamat AlMoallem
- Department of Ophthalmology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
- King Saud University Medical City (KSUMC), Riyadh, Saudi Arabia
| | | |
Collapse
|
18
|
Abstract
The gut microbiome influences many host physiologies, spanning gastrointestinal function, metabolism, immune homeostasis, neuroactivity, and behavior. Many microbial effects on the host are orchestrated by bidirectional interactions between the microbiome and immune system. Imbalances in this dialogue can lead to immune dysfunction and immune-mediated conditions in distal organs including the brain. Dysbiosis of the gut microbiome and dysregulated neuroimmune responses are common comorbidities of neurodevelopmental, neuropsychiatric, and neurological disorders, highlighting the importance of the gut microbiome–neuroimmune axis as a regulator of central nervous system homeostasis. In this review, we discuss recent evidence supporting a role for the gut microbiome in regulating the neuroimmune landscape in health and disease. Expected final online publication date for the Annual Review of Immunology, Volume 40 is April 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Lewis W. Yu
- Department of Integrative Biology and Physiology, University of California, Los Angeles, California 90095, USA;, ,
| | - Gulistan Agirman
- Department of Integrative Biology and Physiology, University of California, Los Angeles, California 90095, USA;, ,
| | - Elaine Y. Hsiao
- Department of Integrative Biology and Physiology, University of California, Los Angeles, California 90095, USA;, ,
| |
Collapse
|
19
|
Khan AS, Wolf A, Langmann T. The AhR ligand 2, 2'-aminophenyl indole (2AI) regulates microglia homeostasis and reduces pro-inflammatory signaling. Biochem Biophys Res Commun 2021; 579:15-21. [PMID: 34583190 DOI: 10.1016/j.bbrc.2021.09.054] [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: 08/12/2021] [Revised: 09/08/2021] [Accepted: 09/21/2021] [Indexed: 10/20/2022]
Abstract
Retinal degeneration is a leading cause of visual impairment and blindness worldwide. Microglia reactivity is a hallmark of neurodegenerative diseases and a driving force for retinal cell death and disease progression. Thus, immunomodulation emerges as a potential therapeutic option. AhR deficiency is known to trigger inflammation and previous studies revealed important roles for AhR ligands in neuroprotection without focusing on microglia. Here, we investigate the anti-inflammatory and antioxidant effects of the synthetic aryl hydrocarbon receptor (AhR) ligand 2, 2'-aminophenyl indole (2AI) on microglia reactivity. We showed that 2AI potently reduced pro-inflammatory gene expression and induced antioxidant genes in activated human and murine microglia cells, in LPS-stimulated retinal explants as well as in stressed human ARPE-19 cells. 2AI also diminished LPS-induced nitric oxide (NO) release, their neurotoxic activity on photoreceptor cells, phagocytosis, and migration in murine BV-2 cells as important functional microglia parameters. siRNA-mediated knockdown of AhR partially prevented the previously observed gene regulatory effects in BV-2 cells. Our results show for the first time, that the synthetic AhR agonist 2AI regulates microglia homeostasis, highlighting AhR as a potential drug target for immunomodulatory and antioxidant therapies.
Collapse
Affiliation(s)
- Amir Saeed Khan
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, 50931, Cologne, Germany.
| | - Anne Wolf
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, 50931, Cologne, Germany.
| | - Thomas Langmann
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, 50931, Cologne, Germany; Center for Molecular Medicine, 50931, Cologne, Germany.
| |
Collapse
|
20
|
Jeong KW. FLII and MLL1 Cooperatively Regulate Aryl Hydrocarbon Receptor-Mediated Transcription in ARPE-19 Cells. Curr Issues Mol Biol 2021; 43:1623-1631. [PMID: 34698116 PMCID: PMC8929146 DOI: 10.3390/cimb43030115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 09/29/2021] [Accepted: 10/06/2021] [Indexed: 11/16/2022] Open
Abstract
Aryl hydrocarbon receptors (AHRs), a class of ligand-dependent nuclear receptors that regulate cellular responses by inducing the expression of various target genes in response to external signals, are implicated in maintaining retinal tissue homeostasis. Previous studies have shown that the regulation of AHR-induced gene expression requires transcriptional co-regulators. However, it is not yet clear how chromatin remodelers, histone methyltransferases and coactivators interact during AHR-mediated gene expression in human retinal cells. In this study, we reveal that the histone methyltransferase MLL1 and the coactivator FLII are involved in AHR-mediated gene expression in retinal pigment epithelial cells. 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) significantly increased the expression of CYP1A1, CYP1B1 and AHRR in ARPE-19 cells, whereas FLII or MLL1 depletion significantly reduced the expression of these genes induced by TCDD. Mechanistically, FLII binds to AHR in a ligand-dependent manner in ARPE-19 cells. In particular, the binding of FLII to MLL1 occurs through the GelB domain of FLII. In addition, MLL1 binds to AHR in a ligand-independent manner. FLII is involved in the recruitment of the BRG1 chromatin remodeler and MLL1 histone methyltransferase to the AHR-regulated CYP1A1 gene region in ARPE-19 cells and consequently, plays an important role in RNA polymerase II binding and transcriptional activity by modulating chromatin accessibility. Our results identify the functions and mechanisms of action of FLII and MLL1 in AHR-induced gene expression in human retinal pigment epithelial cells.
Collapse
Affiliation(s)
- Kwang Won Jeong
- Gachon Research Institute of Pharmaceutical Sciences, College of Pharmacy, Gachon University, 191 Hambakmoero, Yeonsu-gu, Incheon 406-799, Korea
| |
Collapse
|
21
|
Liu YV, Konar G, Aziz K, Tun SBB, Hua CHE, Tan B, Tian J, Luu CD, Barathi VA, Singh MS. Localized Structural and Functional Deficits in a Nonhuman Primate Model of Outer Retinal Atrophy. Invest Ophthalmol Vis Sci 2021; 62:8. [PMID: 34643661 PMCID: PMC8525844 DOI: 10.1167/iovs.62.13.8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Purpose Cell-based therapy development for geographic atrophy (GA) in age-related macular degeneration (AMD) is hampered by the paucity of models of localized photoreceptor and retinal pigment epithelium (RPE) degeneration. We aimed to characterize the structural and functional deficits in a laser-induced nonhuman primate model, including an analysis of the choroid. Methods Macular laser photocoagulation was applied in four macaques. Fundus photography, optical coherence tomography (OCT), dye angiography, and OCT-angiography were conducted over 4.5 months, with histological correlation. Longitudinal changes in spatially resolved macular dysfunction were measured using multifocal electroretinography (MFERG). Results Lesion features, depending on laser settings, included photoreceptor layer degeneration, inner retinal sparing, skip lesions, RPE elevation, and neovascularization. The intralesional choroid was degenerated. The normalized mean MFERG amplitude within lesions was consistently lower than control regions (0.94 ± 0.35 vs. 1.10 ± 0.27, P = 0.032 at month 1, 0.67 ± 0.22 vs. 0.83 ± 0.15, P = 0.0002 at month 2, and 0.97 ± 0.31 vs. 1.20 ± 0.21, P < 0.0001 at month 3.5). The intertest variation of mean MFERG amplitudes in rings 1 to 5 ranged from 13.0% to 26.0% in normal eyes. Conclusions Laser application in this model caused localized outer retinal, RPE, and choriocapillaris loss. Localized dysfunction was apparent by MFERG in the first month after lesion induction. Correlative structure-function testing may be useful for research on the functional effects of stem cell-based therapy for GA. MFERG amplitude data should be interpreted in the context of relatively high intertest variability of the rings that correspond to the central macula. Sustained choroidal insufficiency may limit long-term subretinal graft viability in this model.
Collapse
Affiliation(s)
- Ying V Liu
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Gregory Konar
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Kanza Aziz
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Sai Bo Bo Tun
- Singapore Eye Research Institute, Singapore National Eye Center, Singapore, Singapore
| | - Candice Ho Ee Hua
- Singapore Eye Research Institute, Singapore National Eye Center, Singapore, Singapore
| | - Bingyao Tan
- Singapore Eye Research Institute, Singapore National Eye Center, Singapore, Singapore.,SERI-NTU Advanced Ocular Engineering (STANCE), Singapore, Singapore
| | - Jing Tian
- Department of Biostatistics, Johns Hopkins University Bloomberg School of Public Health, Baltimore, United States
| | - Chi D Luu
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Victoria, Australia.,Ophthalmology, Department of Surgery, University of Melbourne, Victoria, Australia
| | - Veluchamy A Barathi
- Singapore Eye Research Institute, Singapore National Eye Center, Singapore, Singapore.,Academic Clinical Program in Ophthalmology, Duke-NUS Graduate Medical School, Singapore, Singapore.,Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Mandeep S Singh
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| |
Collapse
|
22
|
Riemann S, Kolibabka M, Busch S, Lin J, Hoffmann S, Gretz N, Feng Y, Wohlfart P, Hammes HP. Microglial Activation Is Associated With Vasoprotection in a Rat Model of Inflammatory Retinal Vasoregression. Front Physiol 2021; 12:660164. [PMID: 33981252 PMCID: PMC8107726 DOI: 10.3389/fphys.2021.660164] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 03/16/2021] [Indexed: 02/02/2023] Open
Abstract
Vascular dysfunction and vasoregression are hallmarks of a variety of inflammatory central nervous system disorders and inflammation-related retinal diseases like diabetic retinopathy. Activation of microglia and the humoral innate immune system are contributing factors. Anti-inflammatory approaches have been proposed as therapies for neurovascular diseases, which include the modulation of microglial activation. The present study aimed at investigating the effects of microglial activation by clodronate-coated liposomes on vasoregression in a model of retinal degeneration. Clodronate treatment over 5 weeks led to an increase in activated CD74+ microglia and completely prevented acellular capillaries and pericyte loss. Gene expression analyses indicated that vasoprotection was due to the induction of vasoprotective factors such as Egr1, Stat3, and Ahr while expression of pro-inflammatory genes remained unchanged. We concluded that activated microglia led to a shift toward induction of pleiotropic protective pathways supporting vasoprotection in neurovascular retinal diseases.
Collapse
Affiliation(s)
- Sarah Riemann
- 5th Medical Department, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Matthias Kolibabka
- 5th Medical Department, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Stephanie Busch
- 5th Medical Department, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Jihong Lin
- 5th Medical Department, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Sigrid Hoffmann
- Medical Research Center, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Norbert Gretz
- Medical Research Center, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Yuxi Feng
- Experimental Pharmacology, European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Paulus Wohlfart
- Sanofi Aventis Deutschland GmbH, TA Diabetes R&D, Frankfurt, Germany
| | - Hans-Peter Hammes
- 5th Medical Department, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
23
|
Gao S, Li Y, Bissig D, Cohen ED, Podolsky RH, Childers KL, Vernon G, Chen S, Berkowitz BA, Qian H. Functional regulation of an outer retina hyporeflective band on optical coherence tomography images. Sci Rep 2021; 11:10260. [PMID: 33986362 PMCID: PMC8119672 DOI: 10.1038/s41598-021-89599-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 04/27/2021] [Indexed: 12/30/2022] Open
Abstract
Human and animal retinal optical coherence tomography (OCT) images show a hyporeflective band (HB) between the photoreceptor tip and retinal pigment epithelium layers whose mechanisms are unclear. In mice, HB magnitude and the external limiting membrane-retinal pigment epithelium (ELM-RPE) thickness appear to be dependent on light exposure, which is known to alter photoreceptor mitochondria respiration. Here, we test the hypothesis that these two OCT biomarkers are linked to metabolic activity of the retina. Acetazolamide, which acidifies the subretinal space, had no significant impact on HB magnitude but produced ELM-RPE thinning. Mitochondrial stimulation with 2,4-dinitrophenol reduced both HB magnitude and ELM-RPE thickness in parallel, and also reduced F-actin expression in the same retinal region, but without altering ERG responses. For mice strains with relatively lower (C57BL/6J) or higher (129S6/ev) rod mitochondrial efficacy, light-induced changes in HB magnitude and ELM-RPE thickness were correlated. Humans, analyzed from published data captured with a different protocol, showed a similar light-dark change pattern in HB magnitude as in the mice. Our results indicate that mitochondrial respiration underlies changes in HB magnitude upstream of the pH-sensitive ELM-RPE thickness response. These two distinct OCT biomarkers could be useful indices for non-invasively evaluating photoreceptor mitochondrial metabolic activity.
Collapse
Affiliation(s)
- Shasha Gao
- Department of Ophthalmology, the First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
- Visual Function Core, National Eye Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Yichao Li
- Visual Function Core, National Eye Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - David Bissig
- Department of Neurology, University of California Davis, Sacramento, CA, USA
| | - Ethan D Cohen
- Division of Biomedical Physics, Office of Science and Engineering Labs, Center for Devices and Radiological Health, Food and Drug Administration, Silver Spring, MD, USA
| | - Robert H Podolsky
- Beaumont Research Institute, Beaumont Health, Royal Oak, MI, 48073, USA
| | | | - Gregory Vernon
- Visual Function Core, National Eye Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Sonia Chen
- Visual Function Core, National Eye Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Bruce A Berkowitz
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - Haohua Qian
- Visual Function Core, National Eye Institute, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
24
|
Toma C, De Cillà S, Palumbo A, Garhwal DP, Grossini E. Oxidative and Nitrosative Stress in Age-Related Macular Degeneration: A Review of Their Role in Different Stages of Disease. Antioxidants (Basel) 2021; 10:antiox10050653. [PMID: 33922463 PMCID: PMC8145578 DOI: 10.3390/antiox10050653] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/20/2021] [Accepted: 04/21/2021] [Indexed: 12/14/2022] Open
Abstract
Although the exact pathogenetic mechanisms leading to age-related macular degeneration (AMD) have not been clearly identified, oxidative damage in the retina and choroid due to an imbalance between local oxidants/anti-oxidant systems leading to chronic inflammation could represent the trigger event. Different in vitro and in vivo models have demonstrated the involvement of reactive oxygen species generated in a highly oxidative environment in the development of drusen and retinal pigment epithelium (RPE) changes in the initial pathologic processes of AMD; moreover, recent evidence has highlighted the possible association of oxidative stress and neovascular AMD. Nitric oxide (NO), which is known to play a key role in retinal physiological processes and in the regulation of choroidal blood flow, under pathologic conditions could lead to RPE/photoreceptor degeneration due to the generation of peroxynitrite, a potentially cytotoxic tyrosine-nitrating molecule. Furthermore, the altered expression of the different isoforms of NO synthases could be involved in choroidal microvascular changes leading to neovascularization. The purpose of this review was to investigate the different pathways activated by oxidative/nitrosative stress in the pathogenesis of AMD, focusing on the mechanisms leading to neovascularization and on the possible protective role of anti-vascular endothelial growth factor agents in this context.
Collapse
Affiliation(s)
- Caterina Toma
- Eye Clinic, University Hospital Maggiore Della Carità, 28100 Novara, Italy; (C.T.); (S.D.C.); (A.P.)
| | - Stefano De Cillà
- Eye Clinic, University Hospital Maggiore Della Carità, 28100 Novara, Italy; (C.T.); (S.D.C.); (A.P.)
- Department of Health Sciences, University East Piedmont “A. Avogadro”, 28100 Novara, Italy
| | - Aurelio Palumbo
- Eye Clinic, University Hospital Maggiore Della Carità, 28100 Novara, Italy; (C.T.); (S.D.C.); (A.P.)
| | - Divya Praveen Garhwal
- Laboratory of Physiology and Experimental Surgery, Department of Translational Medicine, University East Piedmont “A. Avogadro”, 28100 Novara, Italy;
| | - Elena Grossini
- Laboratory of Physiology and Experimental Surgery, Department of Translational Medicine, University East Piedmont “A. Avogadro”, 28100 Novara, Italy;
- Correspondence: ; Tel.:+39-0321-660526
| |
Collapse
|
25
|
Innate Immunity in Age-Related Macular Degeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1256:121-141. [PMID: 33848000 DOI: 10.1007/978-3-030-66014-7_5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Multiple lines of investigation have demonstrated that inflammation plays significant roles in etiology of age-related macular degeneration (AMD). Although interventional trials in AMD therapy targeting inflammatory pathways have been conducted, they have not yet been successful and a detailed understanding as to why some have failed is still elusive. One limitation is the relative dearth of information on how immune cells interact with retinal cells to generate AMD phenotypes at each disease stage. Here, we summarize current research evidence and hypotheses regarding potential pathogenic roles of innate immune cells in the eye, which include resident retinal microglia, macrophages derived from infiltrating systemic monocytes, and macrophages resident in the choroid. We relate recent findings regarding the physiology, function, and cellular interactions involving innate immune cells in the retina and choroid to AMD-related processes, including: (1) drusen formation and regression, (2) the onset and spread of degeneration in late atrophic AMD, and (3) the initiation, growth, and exudation of neovascular vessels in late "wet" AMD. Understanding how innate immune cells contribute to specific AMD phenotypes can assist in generating a comprehensive view on the inflammatory etiology of AMD and aid in identifying anti-inflammatory therapeutic strategies and selecting appropriate clinical outcomes for the planned interventions.
Collapse
|
26
|
Soundara Pandi SP, Ratnayaka JA, Lotery AJ, Teeling JL. Progress in developing rodent models of age-related macular degeneration (AMD). Exp Eye Res 2020; 203:108404. [PMID: 33340497 DOI: 10.1016/j.exer.2020.108404] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 12/13/2020] [Accepted: 12/14/2020] [Indexed: 12/25/2022]
Abstract
Age-related macular degeneration (AMD) is the leading cause of irreversible central vision loss, typically affecting individuals from mid-life onwards. Its multifactorial aetiology and the lack of any effective treatments has spurred the development of animal models as research and drug discovery tools. Several rodent models have been developed which recapitulate key features of AMD and provide insights into its underlying pathology. These have contributed to making significant progress in understanding the disease and the identification of novel therapeutic targets. However, a major caveat with existing models is that they do not demonstrate the full disease spectrum. In this review, we outline advances in rodent AMD models from the last decade. These models feature various hallmarks associated with AMD, including oxidative stress, hypoxia, immune dysregulation, genetic mutations and environmental risk factors. The review summarises the methods by which each model was created, its pathological characteristics as well as its relation to the disease in humans.
Collapse
Affiliation(s)
- Sudha Priya Soundara Pandi
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, MP806, Tremona Road, Southampton, SO16 6YD, United Kingdom
| | - J Arjuna Ratnayaka
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, MP806, Tremona Road, Southampton, SO16 6YD, United Kingdom.
| | - Andrew J Lotery
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, MP806, Tremona Road, Southampton, SO16 6YD, United Kingdom; Eye Unit, University Hospital Southampton NHS Foundation Trust, Southampton, SO16 6YD, United Kingdom.
| | - Jessica L Teeling
- Biological Sciences, Faculty of Natural and Environmental Sciences, University of Southampton, MP840, Tremona Road, Southampton, SO16 6YD, United Kingdom.
| |
Collapse
|
27
|
Kim SY, Kambhampati SP, Bhutto IA, McLeod DS, Lutty GA, Kannan RM. Evolution of oxidative stress, inflammation and neovascularization in the choroid and retina in a subretinal lipid induced age-related macular degeneration model. Exp Eye Res 2020; 203:108391. [PMID: 33307075 DOI: 10.1016/j.exer.2020.108391] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 10/16/2020] [Accepted: 12/05/2020] [Indexed: 01/19/2023]
Abstract
Oxidative stress, inflammation and neovascularization are the key pathological events that are implicated in human age-related macular degeneration (AMD). There are a limited number of animal models available for evaluating and developing new therapies. Most models represent late exudative or neovascular AMD (nAMD) but there is a relative paucity of models that mimic early events in AMD. The purpose of this study is to characterize the evolution of oxidative stress, inflammation, retinal degeneration and neovascularization in a rat model of AMD, created by subretinal injection of human lipid hydroperoxide (HpODE) that found in the sub-macular region in aged and AMD patients. Subretinal HpODE induced retinal pigment epithelium (RPE) and retinal degeneration resulting in loss of RPE cells, photoreceptors and retinal thinning. RPE degeneration and atrophy were detected by day 5, followed by neural tissue degeneration at day 12 with robust TUNEL positive cells. Western blot analysis confirmed an increase in pro-apoptotic Bak protein at day 12 in retinal tissues. Oxidative damage biomarkers (4-hydroxynonenal, malondialdehyde, 8-hydroxy-2'-deoxyguanosine, and nitrotyrosine) increased in retinal tissue from days 5-12. Müller glial activation was observed in the HpODE injected area at day 5 followed by its remodeling and migration in the outer retina by day 20. RT-qPCR analysis further indicated upregulation of pro-inflammatory genes (TNF-α, IL-1β and IL-6) both in retinal and RPE/choroidal tissue as early as day 2 and persisted until day 12. Upregulation of oxidative stress markers such as NADPH oxidase (NOX and DOUX family) was detected early in retinal tissue by day 2 followed by its upregulation in choroidal tissue at day 5. Neovascularization was demonstrated from day 12 to day 20 post HpODE injection in choroidal tissue. The results from this study indicate that subretinal HpODE induces advanced AMD phenotypes comprising many aspects of both dry/early and late) and neovascular/late AMD as observed in humans. Within 3 weeks via oxidative damage, upregulation of reactive oxygen species and pro-inflammatory genes, pro-apoptotic Bak and pro-angiogenic VEGF upregulation occurs leading to CNV formation. This experimental model of subretinal HpODE is an appropriate model for the study of AMD and provides an important platform for translational and basic research in developing new therapies particularly for early/dry AMD where currently no viable therapies are available.
Collapse
Affiliation(s)
- Soo-Young Kim
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Siva P Kambhampati
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Imran A Bhutto
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - D Scott McLeod
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Gerard A Lutty
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Rangaramanujam M Kannan
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
28
|
Khan AS, Langmann T. Indole-3-carbinol regulates microglia homeostasis and protects the retina from degeneration. J Neuroinflammation 2020; 17:327. [PMID: 33143743 PMCID: PMC7640677 DOI: 10.1186/s12974-020-01999-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 10/14/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Retinal degenerative diseases significantly contribute to visual impairment and blindness. Microglia reactivity is a hallmark of neurodegenerative diseases including retinal cell death and immunomodulation emerges as a therapeutic option. Indole-3-carbinol (I3C) is a natural ligand of aryl hydrocarbon receptor (AhR), with potent immunomodulatory properties. Here, we hypothesized that I3C may inhibit microglia reactivity and exert neuroprotective effects in the light-damaged murine retina mimicking important immunological aspects of retinal degeneration. METHODS BV-2 microglia were treated in vitro with I3C followed by lipopolysaccharide (LPS) stimulation to analyze pro-inflammatory and anti-oxidant responses by quantitative real-time PCR (qRT-PCR) and Western blots. Nitric oxide (NO) secretion, caspase 3/7 levels, phagocytosis rates, migration, and morphology were analyzed in control and AhR knockdown cells. I3C or vehicle was systemically applied to light-treated BALB/cJ mice as an experimental model of retinal degeneration. Pro-inflammatory and anti-oxidant responses in the retina were examined by qRT-PCR, ELISA, and Western blots. Immunohistochemical staining of retinal flat mounts and cryosections were performed. The retinal thickness and structure were evaluated by in vivo imaging using spectral domain-optical coherence tomography (SD-OCT). RESULTS The in vitro data showed that I3C potently diminished LPS-induced pro-inflammatory gene expression of I-NOS, IL-1ß, NLRP3, IL-6, and CCL2 and induced anti-oxidants gene levels of NQO1, HMOX1, and CAT1 in BV-2 cells. I3C also reduced LPS-induced NO secretion, phagocytosis, and migration as important functional microglia parameters. siRNA-mediated knockdown of AhR partially prevented the previously observed gene regulatory events. The in vivo experiments revealed that I3C treatment diminished light-damage induced I-NOS, IL-1ß, NLRP3, IL-6, and CCL2 transcripts and also reduced CCL2, I-NOS, IL-1ß, p-NFkBp65 protein levels in mice. Moreover, I3C increased anti-oxidant NQO1 and HMOX1 protein levels in light-exposed retinas. Finally, I3C therapy prevented the accumulation of amoeboid microglia in the subretinal space and protected from retinal degeneration. CONCLUSIONS The AhR ligand I3C potently counter-acts microgliosis and light-induced retinal damage, highlighting a potential treatment concept for retinal degeneration.
Collapse
Affiliation(s)
- Amir Saeed Khan
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Joseph-Stelzmann-Str. 9, D-50931, Cologne, Germany
| | - Thomas Langmann
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Joseph-Stelzmann-Str. 9, D-50931, Cologne, Germany.
- Center for Molecular Medicine Cologne, Cologne, Germany.
| |
Collapse
|
29
|
Choudhary M, Malek G. The Aryl Hydrocarbon Receptor: A Mediator and Potential Therapeutic Target for Ocular and Non-Ocular Neurodegenerative Diseases. Int J Mol Sci 2020; 21:ijms21186777. [PMID: 32947781 PMCID: PMC7555571 DOI: 10.3390/ijms21186777] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/10/2020] [Accepted: 09/11/2020] [Indexed: 02/06/2023] Open
Abstract
The aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor, which senses environmental, dietary or metabolic signals to mount a transcriptional response, vital in health and disease. As environmental stimuli and metabolic products have been shown to impact the central nervous system (CNS), a burgeoning area of research has been on the role of the AHR in ocular and non-ocular neurodegenerative diseases. Herein, we summarize our current knowledge, of AHR-controlled cellular processes and their impact on regulating pathobiology of select ocular and neurodegenerative diseases. We catalogue animal models generated to study the role of the AHR in tissue homeostasis and disease pathogenesis. Finally, we discuss the potential of targeting the AHR pathway as a therapeutic strategy, in the context of the maladies of the eye and brain.
Collapse
Affiliation(s)
- Mayur Choudhary
- Department of Ophthalmology, Duke University School of Medicine, 2351 Erwin Road, P.O. Box 3802, Durham, NC 27705, USA
- Correspondence: (M.C.); (G.M.)
| | - Goldis Malek
- Department of Ophthalmology, Duke University School of Medicine, 2351 Erwin Road, P.O. Box 3802, Durham, NC 27705, USA
- Department of Pathology, Duke University School of Medicine, Durham, NC 27705, USA
- Correspondence: (M.C.); (G.M.)
| |
Collapse
|
30
|
Makabe K, Sugita S, Mandai M, Futatsugi Y, Takahashi M. Microglia dynamics in retinitis pigmentosa model: formation of fundus whitening and autofluorescence as an indicator of activity of retinal degeneration. Sci Rep 2020; 10:14700. [PMID: 32895435 PMCID: PMC7477572 DOI: 10.1038/s41598-020-71626-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Accepted: 06/04/2020] [Indexed: 01/04/2023] Open
Abstract
In patients with retinitis pigmentosa (RP), color fundus photography and fundus autofluorescence (FAF) have been used to estimate the disease progression. To understand the origin and the diagnostic interpretation of the fundus color and FAF, we performed in vivo imaging of fundus color and FAF together with histological analyses of the retinal degeneration process using the RP model mice, rd10. FAF partly represented the accumulation of microglia in the photoreceptor outer segments. Fundus whitening suggested the presence of apoptotic cells, which spatiotemporally preceded increase in FAF. We observed two patterns of FAF localization, arcuate and diffuse, each indicating different pattern of apoptosis, wavy and diffuse, respectively. Diffuse pattern of apoptosis was suppressed in dark-raised rd10 mice, in which outer nuclear layer (ONL) loss was significantly suppressed. The occupancy of FAF correlated with the thinning rate of the ONL. Fractalkine, a microglia chemotactic factor, was detected in apoptotic photoreceptors, suggesting chemokine-induced recruitment of microglia into the ONL, which paralleled with accelerated ONL loss and increased FAF occupancy. Thus, we propose that the degree of photoreceptor apoptosis and the rate of ONL thinning in RP patients might be read from the fundus color and the FAF.
Collapse
Affiliation(s)
- Kenichi Makabe
- Laboratory for Retinal Regeneration, Center for Biosystems Dynamics Research, RIKEN, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan.,Department of Ophthalmology, Kobe Kaisei Hospital, 3-11-15 Shinoharakitamachi, Nada-ku, Kobe, 657-0068, Japan
| | - Sunao Sugita
- Laboratory for Retinal Regeneration, Center for Biosystems Dynamics Research, RIKEN, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan.
| | - Michiko Mandai
- Laboratory for Retinal Regeneration, Center for Biosystems Dynamics Research, RIKEN, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan
| | - Yoko Futatsugi
- Laboratory for Retinal Regeneration, Center for Biosystems Dynamics Research, RIKEN, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan
| | - Masayo Takahashi
- Laboratory for Retinal Regeneration, Center for Biosystems Dynamics Research, RIKEN, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan
| |
Collapse
|
31
|
Zhang X, Henneman NF, Girardot PE, Sellers JT, Chrenek MA, Li Y, Wang J, Brenner C, Nickerson JM, Boatright JH. Systemic Treatment With Nicotinamide Riboside Is Protective in a Mouse Model of Light-Induced Retinal Degeneration. Invest Ophthalmol Vis Sci 2020; 61:47. [PMID: 32852543 PMCID: PMC7452859 DOI: 10.1167/iovs.61.10.47] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 07/21/2020] [Indexed: 02/06/2023] Open
Abstract
Purpose Maintaining levels of nicotinamide adenine dinucleotide (NAD+), a coenzyme critical for cellular energetics and biosynthetic pathways, may be therapeutic in retinal disease because retinal NAD+ levels decline during retinal damage and degeneration. The purpose of this study was to investigate whether systemic treatment with nicotinamide riboside (NR), a NAD+ precursor that is orally deliverable and well-tolerated by humans, is protective in a mouse model of light-induced retinal degeneration. Methods Mice were injected intraperitoneally with vehicle or NR the day before and the morning of exposure to degeneration-inducing levels of light. Retinal function was assessed by electroretinography and in vivo retinal morphology and inflammation was assessed by optical coherence tomography. Post mortem retina sections were assessed for morphology, TUNEL, and inflammatory markers Iba1 and GFAP. Retinal NAD+ levels were enzymatically assayed. Results Exposure to degeneration-inducing levels of light suppressed retinal NAD+ levels. Mice undergoing light-induced retinal degeneration exhibited significantly suppressed retinal function, severely disrupted photoreceptor cell layers, and increased apoptosis and inflammation in the outer retina. Treatment with NR increased levels of NAD+ in retina and prevented these deleterious outcomes. Conclusions This study is the first to report the protective effects of NR treatment in a mouse model of retinal degeneration. The positive outcomes, coupled with human tolerance to NR dosing, suggest that maintaining retinal NAD+ via systemic NR treatment should be further explored for clinical relevance.
Collapse
Affiliation(s)
- Xian Zhang
- Department of Ophthalmology, School of Medicine, Emory University, Atlanta, Georgia, United States
- Department of Ophthalmology, Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Nathaniel F. Henneman
- Department of Ophthalmology, School of Medicine, Emory University, Atlanta, Georgia, United States
- Institut Necker-Enfants Malades (INEM), INSERM U1151/CNRS UMR 8253, 75015 Paris, France
- Department of Diabetes & Cancer Metabolism, City of Hope National Medical Center, Duarte, California, United States
| | - Preston E. Girardot
- Department of Ophthalmology, School of Medicine, Emory University, Atlanta, Georgia, United States
| | - Jana T. Sellers
- Department of Ophthalmology, School of Medicine, Emory University, Atlanta, Georgia, United States
| | - Micah A. Chrenek
- Department of Ophthalmology, School of Medicine, Emory University, Atlanta, Georgia, United States
| | - Ying Li
- Department of Ophthalmology, School of Medicine, Emory University, Atlanta, Georgia, United States
| | - Jiaxing Wang
- Department of Ophthalmology, School of Medicine, Emory University, Atlanta, Georgia, United States
| | - Charles Brenner
- Department of Diabetes & Cancer Metabolism, City of Hope National Medical Center, Duarte, California, United States
| | - John M. Nickerson
- Department of Ophthalmology, School of Medicine, Emory University, Atlanta, Georgia, United States
| | - Jeffrey H. Boatright
- Department of Ophthalmology, School of Medicine, Emory University, Atlanta, Georgia, United States
- Center for Visual & Neurocognitive Rehabilitation, Atlanta VAHS, Decatur, Georgia, United States
| |
Collapse
|
32
|
Microbial tryptophan metabolites regulate gut barrier function via the aryl hydrocarbon receptor. Proc Natl Acad Sci U S A 2020; 117:19376-19387. [PMID: 32719140 DOI: 10.1073/pnas.2000047117] [Citation(s) in RCA: 360] [Impact Index Per Article: 72.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Inflammatory bowel diseases (IBDs), including Crohn's disease and ulcerative colitis, are associated with dysbiosis of the gut microbiome. Emerging evidence suggests that small-molecule metabolites derived from bacterial breakdown of a variety of dietary nutrients confer a wide array of host benefits, including amelioration of inflammation in IBDs. Yet, in many cases, the molecular pathways targeted by these molecules remain unknown. Here, we describe roles for three metabolites-indole-3-ethanol, indole-3-pyruvate, and indole-3-aldehyde-which are derived from gut bacterial metabolism of the essential amino acid tryptophan, in regulating intestinal barrier function. We determined that these metabolites protect against increased gut permeability associated with a mouse model of colitis by maintaining the integrity of the apical junctional complex and its associated actin regulatory proteins, including myosin IIA and ezrin, and that these effects are dependent on the aryl hydrocarbon receptor. Our studies provide a deeper understanding of how gut microbial metabolites affect host defense mechanisms and identify candidate pathways for prophylactic and therapeutic treatments for IBDs.
Collapse
|
33
|
Ramos-García NA, Orozco-Ibarra M, Estudillo E, Elizondo G, Gómez Apo E, Chávez Macías LG, Sosa-Ortiz AL, Torres-Ramos MA. Aryl Hydrocarbon Receptor in Post-Mortem Hippocampus and in Serum from Young, Elder, and Alzheimer's Patients. Int J Mol Sci 2020; 21:ijms21061983. [PMID: 32183254 PMCID: PMC7139760 DOI: 10.3390/ijms21061983] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 03/09/2020] [Accepted: 03/10/2020] [Indexed: 11/16/2022] Open
Abstract
One of the characteristics of the cerebral aging process is the presence of chronic inflammation through glial cells, which is particularly significant in neurodegeneration. On the other hand, it has been demonstrated that the aryl hydrocarbon receptor (AHR) participates in the inflammatory response. Currently, evidence in animal models shows that the hallmarks of aging are associated with changes in the AHR levels. However, there is no information concerning the behavior and participation of AHR in the human aging brain or in Alzheimer’s disease (AD). We evaluated the expression of AHR in human hippocampal post-mortem tissue and its association with reactive astrocytes by immunohistochemistry. Besides this, we analyzed through ELISA the AHR levels in blood serum from young and elder participants, and from AD patients. The levels of AHR and glial fibrillar acid protein were higher in elder than in young post-mortem brain samples. AHR was localized mainly in the cytosol of astrocytes and displayed a pattern that resembles extracellular vesicles; this latter feature was more conspicuous in AD subjects. We found higher serum levels of AHR in AD patients than in the other participants. These results suggest that AHR participates in the aging process, and probably in the development of neurodegenerative diseases like AD.
Collapse
Affiliation(s)
- Nicte Alaide Ramos-García
- Unidad Periférica de Neurociencias, Instituto Nacional de Neurología y Neurocirugía/Universidad Nacional Autónoma de México. Av. Insurgentes Sur No. 3877 Col. La Fama, Tlalpan, C.P. 14269, Ciudad de México, Mexico;
- Departamento de Biología Celular, CINVESTAV-IPN, Av. Instituto Politécnico Nacional No. 2508, Col. San Pedro Zacatenco, Gustavo A. Madero, C.P. 07360, Ciudad de México, Mexico;
| | - Marisol Orozco-Ibarra
- Laboratorio de Neurobiología Molecular y Celular, Instituto Nacional de Neurología y Neurocirugía. Av. Insurgentes Sur No. 3877 Col. La Fama, Tlalpan, C.P. 14269, Ciudad de México, Mexico;
| | - Enrique Estudillo
- Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía/Universidad Nacional Autónoma de México. Av. Insurgentes Sur No. 3877 Col. La Fama, Tlalpan, C.P. 14269, Ciudad de México, Mexico;
| | - Guillermo Elizondo
- Departamento de Biología Celular, CINVESTAV-IPN, Av. Instituto Politécnico Nacional No. 2508, Col. San Pedro Zacatenco, Gustavo A. Madero, C.P. 07360, Ciudad de México, Mexico;
| | - Erick Gómez Apo
- Hospital General de México, “Dr. Eduardo Liceaga”. Dr. Balmis No. 148, Col. Doctores, Cuauhtémoc, C.P. 06720, Ciudad de México, Mexico; (E.G.A.); (L.G.C.M.)
| | - Laura Graciela Chávez Macías
- Hospital General de México, “Dr. Eduardo Liceaga”. Dr. Balmis No. 148, Col. Doctores, Cuauhtémoc, C.P. 06720, Ciudad de México, Mexico; (E.G.A.); (L.G.C.M.)
| | - Ana Luisa Sosa-Ortiz
- Laboratorio de Demencias, Instituto Nacional de Neurología y Neurocirugía, Av. Insurgentes Sur No. 3877 Col. La Fama, Tlalpan, C.P. 14269, Ciudad de México, Mexico;
| | - Mónica Adriana Torres-Ramos
- Unidad Periférica de Neurociencias, Instituto Nacional de Neurología y Neurocirugía/Universidad Nacional Autónoma de México. Av. Insurgentes Sur No. 3877 Col. La Fama, Tlalpan, C.P. 14269, Ciudad de México, Mexico;
- Correspondence: ; Tel.: +52-55-56063822 (ext. 3045)
| |
Collapse
|
34
|
Chen D, Chao DL, Rocha L, Kolar M, Nguyen Huu VA, Krawczyk M, Dasyani M, Wang T, Jafari M, Jabari M, Ross KD, Saghatelian A, Hamilton BA, Zhang K, Skowronska‐Krawczyk D. The lipid elongation enzyme ELOVL2 is a molecular regulator of aging in the retina. Aging Cell 2020; 19:e13100. [PMID: 31943697 PMCID: PMC6996962 DOI: 10.1111/acel.13100] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 11/22/2019] [Accepted: 12/12/2019] [Indexed: 12/31/2022] Open
Abstract
Methylation of the regulatory region of the elongation of very-long-chain fatty acids-like 2 (ELOVL2) gene, an enzyme involved in elongation of long-chain polyunsaturated fatty acids, is one of the most robust biomarkers of human age, but the critical question of whether ELOVL2 plays a functional role in molecular aging has not been resolved. Here, we report that Elovl2 regulates age-associated functional and anatomical aging in vivo, focusing on mouse retina, with direct relevance to age-related eye diseases. We show that an age-related decrease in Elovl2 expression is associated with increased DNA methylation of its promoter. Reversal of Elovl2 promoter hypermethylation in vivo through intravitreal injection of 5-Aza-2'-deoxycytidine (5-Aza-dc) leads to increased Elovl2 expression and rescue of age-related decline in visual function. Mice carrying a point mutation C234W that disrupts Elovl2-specific enzymatic activity show electrophysiological characteristics of premature visual decline, as well as early appearance of autofluorescent deposits, well-established markers of aging in the mouse retina. Finally, we find deposits underneath the retinal pigment epithelium in Elovl2 mutant mice, containing components found in human drusen, a pathologic hallmark of age related macular degeneration. These findings indicate that ELOVL2 activity regulates aging in mouse retina, provide a molecular link between polyunsaturated fatty acids elongation and visual function, and suggest novel therapeutic strategies for the treatment of age-related eye diseases.
Collapse
Affiliation(s)
- Daniel Chen
- Shiley Eye InstituteViterbi Family Department of OphthalmologyUniversity of California San DiegoLa JollaCAUSA
| | - Daniel L. Chao
- Shiley Eye InstituteViterbi Family Department of OphthalmologyUniversity of California San DiegoLa JollaCAUSA
| | - Lorena Rocha
- Shiley Eye InstituteViterbi Family Department of OphthalmologyUniversity of California San DiegoLa JollaCAUSA
| | - Matthew Kolar
- The Salk Institute for Biological StudiesClayton Foundation Laboratories for Peptide BiologyLa JollaCAUSA
| | - Viet Anh Nguyen Huu
- Shiley Eye InstituteViterbi Family Department of OphthalmologyUniversity of California San DiegoLa JollaCAUSA
| | - Michal Krawczyk
- Shiley Eye InstituteViterbi Family Department of OphthalmologyUniversity of California San DiegoLa JollaCAUSA
| | - Manish Dasyani
- Shiley Eye InstituteViterbi Family Department of OphthalmologyUniversity of California San DiegoLa JollaCAUSA
| | - Tina Wang
- Department of MedicineUniversity of California San DiegoLa JollaCAUSA
| | - Maryam Jafari
- Shiley Eye InstituteViterbi Family Department of OphthalmologyUniversity of California San DiegoLa JollaCAUSA
| | - Mary Jabari
- Shiley Eye InstituteViterbi Family Department of OphthalmologyUniversity of California San DiegoLa JollaCAUSA
| | - Kevin D. Ross
- Department of Cellular and Molecular MedicineUniversity of California San DiegoLa JollaCAUSA
| | - Alan Saghatelian
- The Salk Institute for Biological StudiesClayton Foundation Laboratories for Peptide BiologyLa JollaCAUSA
| | - Bruce A. Hamilton
- Department of Cellular and Molecular MedicineUniversity of California San DiegoLa JollaCAUSA
- Institute for Genomic MedicineUniversity of California San DiegoLa JollaCAUSA
| | - Kang Zhang
- Shiley Eye InstituteViterbi Family Department of OphthalmologyUniversity of California San DiegoLa JollaCAUSA
| | - Dorota Skowronska‐Krawczyk
- Shiley Eye InstituteViterbi Family Department of OphthalmologyUniversity of California San DiegoLa JollaCAUSA
- Atkinson Laboratory for Regenerative MedicineUniversity of California San DiegoLa JollaCAUSA
| |
Collapse
|
35
|
The aryl hydrocarbon receptor: an environmental sensor integrating immune responses in health and disease. Nat Rev Immunol 2019; 19:184-197. [PMID: 30718831 DOI: 10.1038/s41577-019-0125-8] [Citation(s) in RCA: 762] [Impact Index Per Article: 127.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The environment, diet, microbiota and body's metabolism shape complex biological processes in health and disease. However, our understanding of the molecular pathways involved in these processes is still limited. The aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor that integrates environmental, dietary, microbial and metabolic cues to control complex transcriptional programmes in a ligand-specific, cell-type-specific and context-specific manner. In this Review, we summarize our current knowledge of AHR and the transcriptional programmes it controls in the immune system. Finally, we discuss the role of AHR in autoimmune and neoplastic diseases of the central nervous system, with a special focus on the gut immune system, the gut-brain axis and the therapeutic potential of targeting AHR in neurological disorders.
Collapse
|
36
|
Gambril JA, Sloan KR, Swain TA, Huisingh C, Zarubina AV, Messinger JD, Ach T, Curcio CA. Quantifying Retinal Pigment Epithelium Dysmorphia and Loss of Histologic Autofluorescence in Age-Related Macular Degeneration. Invest Ophthalmol Vis Sci 2019; 60:2481-2493. [PMID: 31173079 PMCID: PMC6557619 DOI: 10.1167/iovs.19-26949] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Purpose Lipofuscin and melanolipofuscin organelles in retinal pigment epithelium (RPE) cells are signal sources for clinical fundus autofluorescence (AF). To elucidate the subcellular basis of AF imaging, we identified, characterized, and quantified the frequency of RPE morphology and AF phenotypes in donor eyes with age-related macular degeneration (AMD). Methods In 25 RPE-Bruch's membrane flat mounts from 25 eyes, we analyzed 0.4-μm z-stack epifluorescence images of RPE stained with phalloidin for actin cytoskeleton. Using a custom ImageJ plugin, we classified cells selected in a systematic unbiased fashion in six phenotypes representing increasing degrees of pathology. For each cell, area, AF intensity, and number of Voronoi neighbors were compared with phenotype 1 (uniform AF, polygonal morphology) via generalized estimating equations. We also analyzed each cell's neighborhood. Results In 29,323 cells, compared with phenotype 1, all other phenotypes, in order of increasing pathology, had significantly larger area, reduced AF, and more variable number of neighbors. Neighborhood area and AF showed similar, but subtler, trends. Cells with highly autofluorescent granule aggregates are no more autofluorescent than others and are in fact lower overall in AF. Pre-aggregates were found in phenotype 1. Phenotype 2, which exhibited degranulation despite normal cytoskeleton, was the most numerous nonhealthy phenotype (16.23%). Conclusions Despite aggregation of granules that created hyperAF aggregates within cells, overall AF on a per cell basis decreased with increasing severity of dysmorphia (abnormal shape). Data motivate further development of subcellular resolution in clinical fundus AF imaging and inform an ongoing reexamination of the role of lipofuscin in AMD.
Collapse
Affiliation(s)
- J Alan Gambril
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham School of Medicine, Birmingham, Alabama, United States
| | - Kenneth R Sloan
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham School of Medicine, Birmingham, Alabama, United States
| | - Thomas A Swain
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham School of Medicine, Birmingham, Alabama, United States
| | - Carrie Huisingh
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham School of Medicine, Birmingham, Alabama, United States
| | - Anna V Zarubina
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham School of Medicine, Birmingham, Alabama, United States
| | - Jeffrey D Messinger
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham School of Medicine, Birmingham, Alabama, United States
| | - Thomas Ach
- Department of Ophthalmology, University Hospital Würzburg, Würzburg, Germany
| | - Christine A Curcio
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham School of Medicine, Birmingham, Alabama, United States
| |
Collapse
|
37
|
Tsai CH, Lee Y, Li CH, Cheng YW, Kang JJ. Down-regulation of aryl hydrocarbon receptor intensifies carcinogen-induced retinal lesion via SOCS3-STAT3 signaling. Cell Biol Toxicol 2019; 36:223-242. [DOI: 10.1007/s10565-019-09499-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 10/16/2019] [Indexed: 11/29/2022]
|
38
|
Blond F, Léveillard T. Functional Genomics of the Retina to Elucidate its Construction and Deconstruction. Int J Mol Sci 2019; 20:E4922. [PMID: 31590277 PMCID: PMC6801968 DOI: 10.3390/ijms20194922] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 10/01/2019] [Indexed: 12/20/2022] Open
Abstract
The retina is the light sensitive part of the eye and nervous tissue that have been used extensively to characterize the function of the central nervous system. The retina has a central position both in fundamental biology and in the physiopathology of neurodegenerative diseases. We address the contribution of functional genomics to the understanding of retinal biology by reviewing key events in their historical perspective as an introduction to major findings that were obtained through the study of the retina using genomics, transcriptomics and proteomics. We illustrate our purpose by showing that most of the genes of interest for retinal development and those involved in inherited retinal degenerations have a restricted expression to the retina and most particularly to photoreceptors cells. We show that the exponential growth of data generated by functional genomics is a future challenge not only in terms of storage but also in terms of accessibility to the scientific community of retinal biologists in the future. Finally, we emphasize on novel perspectives that emerge from the development of redox-proteomics, the new frontier in retinal biology.
Collapse
Affiliation(s)
- Frédéric Blond
- Department of Genetics, Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France.
| | - Thierry Léveillard
- Department of Genetics, Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France.
| |
Collapse
|
39
|
Zhang X, Girardot PE, Sellers JT, Li Y, Wang J, Chrenek MA, Wu W, Skelton H, Nickerson JM, Pardue MT, Boatright JH. Wheel running exercise protects against retinal degeneration in the I307N rhodopsin mouse model of inducible autosomal dominant retinitis pigmentosa. Mol Vis 2019; 25:462-476. [PMID: 31523123 PMCID: PMC6707757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Accepted: 08/19/2019] [Indexed: 10/25/2022] Open
Abstract
Purpose We previously reported that modest running exercise protects photoreceptors in mice undergoing light-induced retinal degeneration and in the rd10 mouse model of autosomal recessive retinitis pigmentosa (arRP). We hypothesized that exercise would protect against other types of retinal degeneration, specifically, in autosomal dominant inherited disease. We tested whether voluntary running wheel exercise is protective in a retinal degeneration mouse model of class B1 autosomal dominant RP (adRP). Methods C57BL/6J mice heterozygous for the mutation in I307N rhodopsin (Rho) (also known as RHOTvrm4/+, or Tvrm4) are normal until exposed to brief but bright light, whereupon rod photoreceptor degeneration ensues. I307N Rho mice were given access to free spinning (active) or locked (inactive) running wheels. Five weeks later, half of each cohort was treated with 0.2% atropine eye drops and exposed to white LED light (6,000 lux) for 5 min, then returned to maintenance housing with wheels. At 1 week or 4 weeks after induction, retinal and visual function was assessed with electroretinogram (ERG) and optomotor response (OMR). In vivo retinal morphology was assessed with optical coherence tomography (OCT), and fundus blue autofluorescence assessed using a scanning laser ophthalmoscope. The mice were then euthanized, and the eyes fixed for paraffin sectioning or flatmounting. The paraffin sections were stained with hematoxylin and eosin (H&E) and terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) to assess retina morphology and apoptosis. Half of the flatmounts were stained for ZO-1 and α-catenin to assess RPE cell structure and stress. (We previously reported that translocation of α-catenin from cell membranes into the cytosol indicates RPE cell stress.) The remaining flatmounts were stained for ZO-1 and Iba-1 to assess the RPE cell size and shape, and inflammatory responses. Results In vivo measures revealed that induction of the I307N Rho degeneration decreased retinal and visual function, decreased the thickness of the retina and photoreceptor layers, and increased the number of blue autofluorescence spots at the level of the photoreceptor-RPE interface. Post-mortem analyses showed that induction caused loss of photoreceptors in the central retinal region, and increased TUNEL labeling in the outer nuclear layer (ONL). The RPE was disrupted 1 week after induction, with changes in cell size and shape accompanied by increased α-catenin translocation and Iba-1 staining. These outcomes were partially but statistically significantly prevented in the exercised mice. The exercised mice that underwent induced I307N Rho degeneration exhibited retinal function and visual function measures that were statistically indistinguishable from that of the uninduced mice, and compared to the unexercised induced mice, had thicker retina and photoreceptor layers, and decreased numbers of subretinal autofluorescent spots. Post-mortem, the retina sections from the exercised mice that had undergone induced I307N Rho degeneration exhibited numbers of photoreceptors that were statistically indistinguishable from those of uninduced mice. Similarly, exercise largely precluded a degeneration-induced increase in TUNEL-positive cells in the ONL. Finally, the RPE of the exercised mice appeared normal, with a regular cell shape and size, and little to no alpha-catenin translocation or Iba-1 immunosignal. Conclusions Voluntary wheel running partially protected against retinal degeneration and inflammation, and RPE disruption in a model of inducible adRP. This is the first report of exercise protection in an adult adRP animal model. It is also the first report of an RPE phenotype in the I307N Rho mouse. These findings add to a growing literature reporting that modest whole-body exercise is protective across a wide range of models of retinal damage and disease, and further highlights the potential for this accessible and inexpensive therapeutic intervention in the ophthalmic clinic.
Collapse
Affiliation(s)
- Xian Zhang
- Department of Ophthalmology, School of Medicine, Emory University, Atlanta, GA,Department of Ophthalmology, Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Preston E. Girardot
- Department of Ophthalmology, School of Medicine, Emory University, Atlanta, GA
| | - Jana T. Sellers
- Department of Ophthalmology, School of Medicine, Emory University, Atlanta, GA
| | - Ying Li
- Department of Ophthalmology, School of Medicine, Emory University, Atlanta, GA
| | - Jiaxing Wang
- Department of Ophthalmology, School of Medicine, Emory University, Atlanta, GA
| | - Micah A. Chrenek
- Department of Ophthalmology, School of Medicine, Emory University, Atlanta, GA
| | - Wenfei Wu
- Department of Ophthalmology, School of Medicine, Emory University, Atlanta, GA,The First Affiliated Hospital of Medical School of Xi’an Jiaotong University, Xi’an, Shan’xi, China
| | | | - John M. Nickerson
- Department of Ophthalmology, School of Medicine, Emory University, Atlanta, GA
| | - Machelle T. Pardue
- Department of Ophthalmology, School of Medicine, Emory University, Atlanta, GA,Center for Visual and Neurocognitive Rehabilitation, Atlanta Veterans Administration Health Care System, Decatur, GA,Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA
| | - Jeffrey H. Boatright
- Department of Ophthalmology, School of Medicine, Emory University, Atlanta, GA,Center for Visual and Neurocognitive Rehabilitation, Atlanta Veterans Administration Health Care System, Decatur, GA
| |
Collapse
|
40
|
Aryl hydrocarbon receptor is indispensable for β-naphthoflavone-induced novel food avoidance and may be involved in LiCl-triggered conditioned taste aversion in rats. Physiol Behav 2019; 204:58-64. [DOI: 10.1016/j.physbeh.2019.02.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 01/18/2019] [Accepted: 02/11/2019] [Indexed: 12/31/2022]
|
41
|
Zhou Y, Li S, Huang L, Yang Y, Zhang L, Yang M, Liu W, Ramasamy K, Jiang Z, Sundaresan P, Zhu X, Yang Z. A splicing mutation in aryl hydrocarbon receptor associated with retinitis pigmentosa. Hum Mol Genet 2019; 27:2563-2572. [PMID: 29726989 DOI: 10.1093/hmg/ddy165] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 04/30/2018] [Indexed: 12/14/2022] Open
Abstract
Retinitis pigmentosa (RP) refers to a group of retinal degenerative diseases, which often lead to vision loss. Although 70 genes have been identified in RP patients, the genetic cause of approximately 30% of RP cases remains unknown. We aimed to identify the cause of the disease in a cohort of RP families by whole exome sequencing. A rare homozygous splicing variant, c.1160 + 1G>A, which introduced skipping of exon 9 of the aryl hydrocarbon receptor (AHR), was identified in family RD-134. This variant is very rare in several exome databases and leads to skipping of exon 9 in the transcript. AHR is expressed in the human retina and is a ligand-activated transcription factor with multiple functions. Mutant AHR failed to promote 2, 3, 7, 8-tetrachlorodibenzo-p-dioxin (TCDD)-induced xenobiotic responsive element (XRE) luciferase activity. In parallel, mutation in AHR abolished activation of its downstream target gene, such as CYP1A1 and CYP1A2. To investigate the in vivo roles of Ahr in the retina, we generated a retina-specific conditional knockout mouse model of Ahr. Comparing with wild-type mouse, Ahr knockout mice exhibited reduced electroretinogram responses at 9 months of age. Retinal histology revealed retinal histology showed the degeneration of photoreceptors with a thinner outer nuclear layer. Thus, our data demonstrate that AHR is associated with RP.
Collapse
Affiliation(s)
- Yu Zhou
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China.,Department of Laboratory Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
| | - Shujin Li
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China.,Chengdu Institute of Biology, Sichuan Translational Medicine Research Hospital, Chinese Academy of Sciences, Chengdu, Sichuan, China
| | - Lulin Huang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China.,Department of Laboratory Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
| | - Yeming Yang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China.,Department of Laboratory Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
| | - Lin Zhang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China.,Department of Laboratory Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
| | - Mu Yang
- Chengdu Institute of Biology, Sichuan Translational Medicine Research Hospital, Chinese Academy of Sciences, Chengdu, Sichuan, China
| | - Wenjing Liu
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Kim Ramasamy
- Retina-Vitreous Services, Aravind Eye Hospital, Madurai, Tamil Nadu, India
| | - Zhilin Jiang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China.,Department of Laboratory Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
| | - Periasamy Sundaresan
- Department of Genetics, Aravind Medical Research Foundation, Aravind Eye Hospital, Madurai, Tamil Nadu, India
| | - Xianjun Zhu
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China.,Chengdu Institute of Biology, Sichuan Translational Medicine Research Hospital, Chinese Academy of Sciences, Chengdu, Sichuan, China.,Department of Laboratory Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China.,Institute of Laboratory Animal Sciences, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
| | - Zhenglin Yang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China.,Chengdu Institute of Biology, Sichuan Translational Medicine Research Hospital, Chinese Academy of Sciences, Chengdu, Sichuan, China.,Department of Laboratory Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
| |
Collapse
|
42
|
Dhingra A, Bell BA, Peachey NS, Daniele LL, Reyes-Reveles J, Sharp RC, Jun B, Bazan NG, Sparrow JR, Kim HJ, Philp NJ, Boesze-Battaglia K. Microtubule-Associated Protein 1 Light Chain 3B, (LC3B) Is Necessary to Maintain Lipid-Mediated Homeostasis in the Retinal Pigment Epithelium. Front Cell Neurosci 2018; 12:351. [PMID: 30349463 PMCID: PMC6186781 DOI: 10.3389/fncel.2018.00351] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 09/19/2018] [Indexed: 12/25/2022] Open
Abstract
Like other neurons, retinal cells utilize autophagic pathways to maintain cell homeostasis. The mammalian retina relies on heterophagy and selective autophagy to efficiently degrade and metabolize ingested lipids with disruption in autophagy associated degradation contributing to age related retinal disorders. The retinal pigment epithelium (RPE) supports photoreceptor cell renewal by daily phagocytosis of shed photoreceptor outer segments (OS). The daily ingestion of these lipid-rich OS imposes a constant degradative burden on these terminally differentiated cells. These cells rely on Microtubule-Associated Protein 1 Light Chain 3 (LC3) family of proteins for phagocytic clearance of the ingested OS. The LC3 family comprises of three highly homologous members, MAP1LC3A (LC3A), MAP1LC3B (LC3B), and MAP1LC3C (LC3C). The purpose of this study was to determine whether the LC3B isoform plays a specific role in maintaining RPE lipid homeostasis. We examined the RPE and retina of the LC3B-/- mouse as a function of age using in vivo ocular imaging and electroretinography coupled with ex vivo, lipidomic analyses of lipid mediators, assessment of bisretinoids as well as imaging of lipid aggregates. Deletion of LC3B resulted in defects within the RPE including increased phagosome accumulation, decreased fatty acid oxidation and a subsequent increase in RPE and sub-RPE lipid deposits. Age-dependent RPE changes included elevated levels of oxidized cholesterol, deposition of 4-HNE lipid peroxidation products, bisretinoid lipofuscin accumulation, and subretinal migration of microglia, collectively likely contributing to loss of retinal function. These observations are consistent with a critical role for LC3B-dependent processes in the maintenance of normal lipid homeostasis in the aging RPE, and suggest that LC3 isoform specific disruption in autophagic processes contribute to AMD-like pathogenesis.
Collapse
Affiliation(s)
- Anuradha Dhingra
- Department of Biochemistry, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Brent A Bell
- Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA, United States
| | - Neal S Peachey
- Cole Eye Institute, Cleveland Clinic, Cleveland, OH, United States.,Louis Stokes Cleveland VA Medical Center, Cleveland, OH, United States.,Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Lauren L Daniele
- Department of Biochemistry, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Juan Reyes-Reveles
- Department of Biochemistry, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Rachel C Sharp
- Department of Biochemistry, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Bokkyoo Jun
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA, United States
| | - Nicolas G Bazan
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA, United States
| | - Janet R Sparrow
- Department of Ophthalmology, Columbia University Medical Center, New York, NY, United States
| | - Hye Jin Kim
- Department of Ophthalmology, Columbia University Medical Center, New York, NY, United States
| | - Nancy J Philp
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Kathleen Boesze-Battaglia
- Department of Biochemistry, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
43
|
Rannug A, Rannug U. The tryptophan derivative 6-formylindolo[3,2-b]carbazole, FICZ, a dynamic mediator of endogenous aryl hydrocarbon receptor signaling, balances cell growth and differentiation. Crit Rev Toxicol 2018; 48:555-574. [PMID: 30226107 DOI: 10.1080/10408444.2018.1493086] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The aryl hydrocarbon receptor (AHR) is not essential to survival, but does act as a key regulator of many normal physiological events. The role of this receptor in toxicological processes has been studied extensively, primarily employing the high-affinity ligand 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). However, regulation of physiological responses by endogenous AHR ligands remains to be elucidated. Here, we review developments in this field, with a focus on 6-formylindolo[3,2-b]carbazole (FICZ), the endogenous ligand with the highest affinity to the receptor reported to date. The binding of FICZ to different isoforms of the AHR seems to be evolutionarily well conserved and there is a feedback loop that controls AHR activity through metabolic degradation of FICZ via the highly inducible cytochrome P450 1A1. Several investigations provide strong evidence that FICZ plays a critical role in normal physiological processes and can ameliorate immune diseases with remarkable efficiency. Low levels of FICZ are pro-inflammatory, providing resistance to pathogenic bacteria, stimulating the anti-tumor functions, and promoting the differentiation of cancer cells by repressing genes in cancer stem cells. In contrast, at high concentrations FICZ behaves in a manner similar to TCDD, exhibiting toxicity toward fish and bird embryos, immune suppression, and activation of cancer progression. The findings are indicative of a dual role for endogenously activated AHR in barrier tissues, aiding clearance of infections and suppressing immunity to terminate a vicious cycle that might otherwise lead to disease. There is not much support for the AHR ligand-specific immune responses proposed, the differences between FICZ and TCDD in this context appear to be explained by the rapid metabolism of FICZ.
Collapse
Affiliation(s)
- Agneta Rannug
- a Karolinska Institutet, Institute of Environmental Medicine , Stockholm , Sweden
| | - Ulf Rannug
- b Department of Molecular Biosciences , The Wenner-Gren Institute, Stockholm University , Stockholm , Sweden
| |
Collapse
|
44
|
The Aryl Hydrocarbon Receptor and the Nervous System. Int J Mol Sci 2018; 19:ijms19092504. [PMID: 30149528 PMCID: PMC6163841 DOI: 10.3390/ijms19092504] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 08/17/2018] [Accepted: 08/21/2018] [Indexed: 12/12/2022] Open
Abstract
The aryl hydrocarbon receptor (or AhR) is a cytoplasmic receptor of pollutants. It translocates into the nucleus upon binding to its ligands, and forms a heterodimer with ARNT (AhR nuclear translocator). The heterodimer is a transcription factor, which regulates the transcription of xenobiotic metabolizing enzymes. Expressed in many cells in vertebrates, it is mostly present in neuronal cell types in invertebrates, where it regulates dendritic morphology or feeding behavior. Surprisingly, few investigations have been conducted to unravel the function of the AhR in the central or peripheral nervous systems of vertebrates. In this review, we will present how the AhR regulates neural functions in both invertebrates and vertebrates as deduced mainly from the effects of xenobiotics. We will introduce some of the molecular mechanisms triggered by the well-known AhR ligand, 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), which impact on neuronal proliferation, differentiation, and survival. Finally, we will point out the common features found in mice that are exposed to pollutants, and in AhR knockout mice.
Collapse
|
45
|
Huang Y, He J, Liang H, Hu K, Jiang S, Yang L, Mei S, Zhu X, Yu J, Kijlstra A, Yang P, Hou S. Aryl Hydrocarbon Receptor Regulates Apoptosis and Inflammation in a Murine Model of Experimental Autoimmune Uveitis. Front Immunol 2018; 9:1713. [PMID: 30090104 PMCID: PMC6068235 DOI: 10.3389/fimmu.2018.01713] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 07/12/2018] [Indexed: 12/14/2022] Open
Abstract
Uveitis is characterized as a common cause of blindness worldwide. Aryl hydrocarbon receptor (AhR), a ligand-activated nuclear receptor, has been implicated to play a role in human uveitis, although the exact mechanisms remain poorly understood. The purpose of this study was to enhance our knowledge concerning the role of AhR during intraocular inflammation. We immunized wild-type and AhR-knockout C57BL/6J mice with IRBP651–670 to induce experimental autoimmune uveitis (EAU). Disease severity was evaluated with both clinical and histopathological grading. Blood–retinal barrier (BRB) integrity was tested by Evans blue and tight junction proteins qualifications. Apoptosis was measured using TdT-mediated dUTP nick end labeling staining. Macrophage/microglia activation and polarization were studied by immunofluorescence and Western blot. Following EAU induction, AhR−/− mice had more severe clinical and histopathological manifestations of uveitis than AhR+/+ mice. Increased vascular permeability and apoptotic cells were observed in AhR−/− EAU mice when compared with AhR+/+ EAU mice. In addition, AhR−/− EAU mice showed evidence of a significantly increased macrophage/microglia cells and a stronger polarization from the M2 to the M1 phenotype as compared to AhR+/+ EAU mice. The levels of pro-inflammatory cytokines including tumor necrosis factor-α (TNF-α), interleukin (IL)-6, and IL-1β were increased in AhR−/− EAU mice, which was associated with the activation of NF-κB and signal transducers and activators of transcription (STAT) pathways. 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), an agonist of AhR, caused a significant decrease in the clinical and histopathological manifestations, preserved BRB integrity, reduced apoptotic cells, inhibited macrophage/microglia activation, and shifted their polarization from M1 toward M2. Moreover, decreased expression of pro-inflammatory cytokines including TNF-α, IL-6, and IL-1β and inhibition of NF-κB and STAT pathways were found in EAU mice following TCDD treatment. In conclusion, AhR activation with TCDD exhibits an immunomodulatory effect by reducing BRB breakdown, inhibiting retinal cell apoptosis, and reducing pro-inflammatory cytokine expression during EAU. The underlying mechanism may involve the modulation of macrophages/microglia polarization and the downregulation of NF-κB and STAT pathways.
Collapse
Affiliation(s)
- Yike Huang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Eye Institute, Chongqing Key Laboratory of Ophthalmology, Chongqing, China
| | - Junchi He
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Eye Institute, Chongqing Key Laboratory of Ophthalmology, Chongqing, China
| | - Huaping Liang
- State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Ke Hu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Eye Institute, Chongqing Key Laboratory of Ophthalmology, Chongqing, China
| | - Shaoqiu Jiang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Eye Institute, Chongqing Key Laboratory of Ophthalmology, Chongqing, China
| | - Lu Yang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Eye Institute, Chongqing Key Laboratory of Ophthalmology, Chongqing, China
| | - Suyin Mei
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Eye Institute, Chongqing Key Laboratory of Ophthalmology, Chongqing, China
| | - Xiao Zhu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Dongguan Scientific Research Center, Guangdong Medical University, Dongguan, China
| | - Jing Yu
- State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Aize Kijlstra
- University Eye Clinic Maastricht, Maastricht, Netherlands
| | - Peizeng Yang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Eye Institute, Chongqing Key Laboratory of Ophthalmology, Chongqing, China
| | - Shengping Hou
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Eye Institute, Chongqing Key Laboratory of Ophthalmology, Chongqing, China
| |
Collapse
|
46
|
Microglia inhibit photoreceptor cell death and regulate immune cell infiltration in response to retinal detachment. Proc Natl Acad Sci U S A 2018; 115:E6264-E6273. [PMID: 29915052 PMCID: PMC6142210 DOI: 10.1073/pnas.1719601115] [Citation(s) in RCA: 116] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Photoreceptor cell death resulting from retinal detachment (RD) causes significant visual loss. While the immune system is activated during RD, its role is still unclear. Microglia are resident immune cells in the retina and are thought to be either protective or deleterious in response to neuronal injury, suggesting context-dependent effects. Here, we demonstrate that microglia limit retinal damage during acute injury, since microglial ablation led to increased photoreceptor death. Microglial morphological–activation changes triggered their migration into injured tissue where they formed intimate connections with infiltrating immune cells and phagocytized injured photoreceptors. These findings provide insight into the microglial response and function during RD, indicating microglia promote photoreceptor survival during acute phase injury by removing potentially damaging cell debris. Retinal detachment (RD) is a sight-threatening complication common in many highly prevalent retinal disorders. RD rapidly leads to photoreceptor cell death beginning within 12 h following detachment. In patients with sustained RD, progressive visual decline due to photoreceptor cell death is common, leading to significant and permanent loss of vision. Microglia are the resident immune cells of the central nervous system, including the retina, and function in the homeostatic maintenance of the neuro-retinal microenvironment. It is known that microglia become activated and change their morphology in retinal diseases. However, the function of activated microglia in RD is incompletely understood, in part because of the lack of microglia-specific markers. Here, using the newly identified microglia marker P2ry12 and microglial depletion strategies, we demonstrate that retinal microglia are rapidly activated in response to RD and migrate into the injured area within 24 h post-RD, where they closely associate with infiltrating macrophages, a population distinct from microglia. Once in the injured photoreceptor layer, activated microglia can be observed to contain autofluorescence within their cell bodies, suggesting they function to phagocytose injured or dying photoreceptors. Depletion of retinal microglia results in increased disease severity and inhibition of macrophage infiltration, suggesting that microglia are involved in regulating neuroinflammation in the retina. Our work identifies that microglia mediate photoreceptor survival in RD and suggests that this effect may be due to microglial regulation of immune cells and photoreceptor phagocytosis.
Collapse
|
47
|
Malek G, Busik J, Grant MB, Choudhary M. Models of retinal diseases and their applicability in drug discovery. Expert Opin Drug Discov 2018; 13:359-377. [PMID: 29382242 DOI: 10.1080/17460441.2018.1430136] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION The impact of vision debilitating diseases is a global public health concern, which will continue until effective preventative and management protocols are developed. Two retinal diseases responsible for the majority of vision loss in the working age adults and elderly populations are diabetic retinopathy (DR) and age-related macular degeneration (AMD), respectively. Model systems, which recapitulate aspects of human pathology, are valid experimental modalities that have contributed to the identification of signaling pathways involved in disease development and consequently potential therapies. Areas covered: The pathology of DR and AMD, which serve as the basis for designing appropriate models of disease, is discussed. The authors also review in vitro and in vivo models of DR and AMD and evaluate the utility of these models in exploratory and pre-clinical studies. Expert opinion: The complex nature of non-Mendelian diseases such as DR and AMD has made identification of effective therapeutic treatments challenging. However, the authors believe that while in vivo models are often criticized for not being a 'perfect' recapitulation of disease, they have been valuable experimentally when used with consideration of the strengths and limitations of the experimental model selected and have a place in the drug discovery process.
Collapse
Affiliation(s)
- Goldis Malek
- a Department of Ophthalmology , Duke University School of Medicine , Durham , NC , USA.,b Department of Pathology , Duke University School of Medicine , Durham , NC , USA
| | - Julia Busik
- c Department of Physiology , Michigan State University , East Lansing , MI , USA
| | - Maria B Grant
- d Department of Ophthalmology , University of Alabama at Birmingham , Birmingham , Al , USA
| | - Mayur Choudhary
- a Department of Ophthalmology , Duke University School of Medicine , Durham , NC , USA
| |
Collapse
|
48
|
Kim J, Jin HL, Jang DS, Jeong KW, Choung SY. Hyperoside (quercetin-3-O-β-D-galactopyranoside) protects A2E-laden retinal pigmented epithelium cells against UVA and blue light-induced apoptosis in vitro and in vivo. J Funct Foods 2018. [DOI: 10.1016/j.jff.2017.11.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
49
|
Loss of NR2E3 represses AHR by LSD1 reprogramming, is associated with poor prognosis in liver cancer. Sci Rep 2017; 7:10662. [PMID: 28878246 PMCID: PMC5587550 DOI: 10.1038/s41598-017-11106-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 08/18/2017] [Indexed: 11/15/2022] Open
Abstract
The aryl hydrocarbon receptor (AHR) plays crucial roles in inflammation, metabolic disorder, and cancer. However, the molecular mechanisms regulating AHR expression remain unknown. Here, we found that an orphan nuclear NR2E3 maintains AHR expression, and forms an active transcriptional complex with transcription factor Sp1 and coactivator GRIP1 in MCF-7 human breast and HepG2 liver cancer cell lines. NR2E3 loss promotes the recruitment of LSD1, a histone demethylase of histone 3 lysine 4 di-methylation (H3K4me2), to the AHR gene promoter region, resulting in repression of AHR expression. AHR expression and responsiveness along with H3K4me2 were significantly reduced in the livers of Nr2e3rd7 (Rd7) mice that express low NR2E3 relative to the livers of wild-type mice. SP2509, an LSD1 inhibitor, fully restored AHR expression and H3K4me2 levels in Rd7 mice. Lastly, we demonstrated that both AHR and NR2E3 are significantly associated with good clinical outcomes in liver cancer. Together, our results reveal a novel link between NR2E3, AHR, and liver cancer via LSD1-mediated H3K4me2 histone modification in liver cancer development.
Collapse
|
50
|
Bennis A, ten Brink JB, Moerland PD, Heine VM, Bergen AA. Comparative gene expression study and pathway analysis of the human iris- and the retinal pigment epithelium. PLoS One 2017; 12:e0182983. [PMID: 28827822 PMCID: PMC5565104 DOI: 10.1371/journal.pone.0182983] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 07/27/2017] [Indexed: 11/25/2022] Open
Abstract
Background The retinal pigment epithelium (RPE) is a neural monolayer lining the back of the eye. Degeneration of the RPE leads to severe vision loss in, so far incurable, diseases such as age-related macular degeneration and some forms of retinitis pigmentosa. A promising future replacement therapy may be autologous iris epithelial cell transdifferentiation into RPE in vitro and, subsequently, transplantation. In this study we compared the gene expression profiles of the iris epithelium (IE) and the RPE. Methods We collected both primary RPE- and IE cells from 5 freshly frozen human donor eyes, using respectively laser dissection microscopy and excision. We performed whole-genome expression profiling using 44k Agilent human microarrays. We investigated the gene expression profiles on both gene and functional network level, using R and the knowledge database Ingenuity. Results The major molecular pathways related to the RPE and IE were quite similar and yielded basic neuro-epithelial cell functions. Nonetheless, we also found major specific differences: For example, genes and molecular pathways, related to the visual cycle and retinol biosynthesis are significantly higher expressed in the RPE than in the IE. Interestingly, Wnt and aryl hydrocarbon receptor (AhR-) signaling pathways are much higher expressed in the IE than in the RPE, suggesting, respectively, a possible pluripotent and high detoxification state of the IE. Conclusions This study provides a valuation of the similarities and differences between the expression profiles of the RPE and IE. Our data combined with that of the literature, represent a most comprehensive perspective on transcriptional variation, which may support future research in the development of therapeutic transplantation of IE.
Collapse
Affiliation(s)
- Anna Bennis
- Department of Clinical Genetics, Academic Medical Center, Amsterdam, The Netherlands
- The Netherlands Institute for Neuroscience (NIN-KNAW), Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - Jacoline B. ten Brink
- Department of Clinical Genetics, Academic Medical Center, Amsterdam, The Netherlands
| | - Perry D. Moerland
- Bioinformatics Laboratory, Department of Clinical Epidemiology, Biostatistics and Bioinformatics, Academic Medical Center, Amsterdam, The Netherlands
| | - Vivi M. Heine
- Department of Pediatrics / Child Neurology, Neuroscience Campus Amsterdam, VU University Medical Centre, Amsterdam, The Netherlands
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit, Amsterdam, The Netherlands
| | - Arthur A. Bergen
- Department of Clinical Genetics, Academic Medical Center, Amsterdam, The Netherlands
- The Netherlands Institute for Neuroscience (NIN-KNAW), Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
- Department of Ophthalmology, Academic Medical Centre, Amsterdam, The Netherlands
- * E-mail:
| |
Collapse
|