1
|
Butovsky O, Rosenzweig N. Alzheimer's disease and age-related macular degeneration: Shared and distinct immune mechanisms. Immunity 2025; 58:1120-1139. [PMID: 40324382 DOI: 10.1016/j.immuni.2025.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 04/09/2025] [Accepted: 04/10/2025] [Indexed: 05/07/2025]
Abstract
Alzheimer's disease (AD) and age-related macular degeneration (AMD) represent the leading causes of dementia and vision impairment in the elderly, respectively. The retina is an extension of the brain, yet these two central nervous system (CNS) compartments are often studied separately. Despite affecting cognition vs. vision, AD and AMD share neuroinflammatory pathways. By comparing these diseases, we can identify converging immune mechanisms and potential cross-applicable therapies. Here, we review immune mechanisms highlighting the shared and distinct aspects of these two age-related neurodegenerative conditions, focusing on responses to hallmark disease manifestations, the opposite role of overlapping immune risk loci, and potential unified therapeutic approaches. We also discuss unique tissue requirements that may dictate different outcomes of conserved immune mechanisms and how we can reciprocally utilize lessons from AD therapeutics to AMD. Looking forward, we suggest promising directions for research, including the exploration of regenerative medicine, gene therapies, and innovative diagnostics.
Collapse
Affiliation(s)
- Oleg Butovsky
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| | - Neta Rosenzweig
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
2
|
Yang X, Zhang Y, Zhou Y, Liu M, Zhao H, Yang Y, Su J. CaMK2A/CREB pathway activation is associated with enhanced mitophagy and neuronal apoptosis in diabetic retinopathy. Sci Rep 2025; 15:12516. [PMID: 40216954 PMCID: PMC11992012 DOI: 10.1038/s41598-025-97371-y] [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: 07/31/2024] [Accepted: 04/03/2025] [Indexed: 04/14/2025] Open
Abstract
Diabetic retinopathy (DR) is a common complication of diabetes mellitus, characterized by progressive neurodegeneration and vision impairment. The Ca2+/calmodulin-dependent protein kinase II alpha (CaMK2A) and cAMP response element-binding protein (CREB) signaling pathway has been implicated in various neurological disorders. However, its role in DR pathogenesis remains elusive. We established a DR mouse model by streptozotocin administration and performed histological, biochemical, and molecular analyses to investigate the involvement of CaMK2A/CREB signaling and its interplay with mitophagy. Additionally, we employed in vitro high-glucose (HG) treatment in primary mouse retinal ganglion cells to dissect the underlying mechanisms. Pharmacological and genetic modulations were utilized to target CaMK2A/CREB pathway and mitophagy. In the DR model, we observed retinal degeneration, increased apoptosis, and reduced neurotransmitter production, accompanied by enhanced mitophagy and activation of the CaMK2A/CREB pathway. HG induction in retinal ganglion cells recapitulated these findings, and autophagy inhibition partially rescued cell death but failed to suppress CaMK2A/CREB activation, suggesting mitophagy as a downstream consequence. CaMK2A knockdown or CREB phosphorylation inhibition attenuated HG-induced mitophagy, apoptosis, and neurotransmitter depletion, while CREB activation exacerbated these effects. CaMK2A silencing mitigated DR progression, oxidative stress, inflammation, and neuronal loss, akin to dopamine/carbidopa administration in DR mouse model. Our findings reveal the involvement of CaMK2A/CREB signaling activation and enhanced mitophagy in DR, suggesting these pathways may be therapeutically relevant targets for DR management.
Collapse
Affiliation(s)
- Xiaochun Yang
- The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, China.
| | - Yuxin Zhang
- The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, China
| | - Yikun Zhou
- The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, China
| | - Mingzhi Liu
- The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, China
| | - Haiyan Zhao
- The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, China
| | - Yang Yang
- The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, China
| | - Jianyun Su
- The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, China
| |
Collapse
|
3
|
Kumar GS, Chidambaranathan GP, Palakkan AA. A Non-destructive, Image Analysis Method for Evaluating Pigmentation in Induced Pluripotent Stem Cell-Derived Retinal Pigment Epithelial Cells. Methods Mol Biol 2025; 2924:235-248. [PMID: 40307647 DOI: 10.1007/978-1-0716-4530-7_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2025]
Abstract
We have developed an image J macro to analyze pigmentation level in iPSC-derived retinal pigment epithelial cells from bright-field images. Human iPSC-derived retinal pigment epithelial cells were treated with hydroquinone to induce hyperpigmentation, and untreated cells were used as controls for the study. The Image analysis results were validated by estimating melanin spectrophotometrically, after lysing the cells. The developed image analysis method will be useful for screening drugs/compounds that induce hyper/hypo pigmentation in cultured iPSC-derived RPE cells.
Collapse
Affiliation(s)
- Gopika S Kumar
- Immunology & Stem Cell Biology, Aravind Medical Research Foundation, Madurai, Tamil Nadu, India
| | | | - Anwar A Palakkan
- Immunology & Stem Cell Biology, Aravind Medical Research Foundation, Madurai, Tamil Nadu, India.
| |
Collapse
|
4
|
Schloesser L, Klose SM, Mauschitz MM, Abdullah Z, Finger RP. The role of immune modulators in age-related macular degeneration. Surv Ophthalmol 2024; 69:851-869. [PMID: 39097172 DOI: 10.1016/j.survophthal.2024.07.009] [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: 03/25/2024] [Revised: 07/27/2024] [Accepted: 07/29/2024] [Indexed: 08/05/2024]
Abstract
We provide an overview of the expanding literature on the role of cytokines and immune mediators in pathophysiology of age-related macular degeneration (AMD). Although many immunological mediators have been linked to AMD pathophysiology, the broader mechanistic picture remains unclear with substantial variations in the levels of evidence supporting these mediators. Therefore, we reviewed the literature considering the varying levels of supporting evidence. A Medical Subject Headings (MeSH) term-based literature research was conducted in September, 2023, consisting of the MeSH terms "cytokine" and "Age-related macular degeneration" connected by the operator "AND". After screening the publications by title, abstract, and full text, a total of 146 publications were included. The proinflammatory cytokines IL-1β (especially in basic research studies), IL-6, IL-8, IL-18, TNF-α, and MCP-1 are the most extensively characterised cytokines/chemokines, highlighting the role of local inflammasome activation and altered macrophage function in the AMD pathophysiology. Among the antiinflammatory mediators IL-4, IL-10, and TGF-β were found to be the most extensively characterised, with IL-4 driving and IL-10 and TGF-β suppressing disease progression. Despite the extensive literature on this topic, a profound understanding of AMD pathophysiology has not yet been achieved. Therefore, further studies are needed to identify potential therapeutic targets, followed by clinical studies.
Collapse
Affiliation(s)
- Lukas Schloesser
- Department of Ophthalmology, University of Bonn, Bonn, Germany; Department of Ophthalmology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
| | - Sara M Klose
- Institute of Molecular Medicine and Experimental Immunology, University of Bonn, Bonn, Germany; Asia-Pacific Centre for Animal Health, Faculty of Science, University of Melbourne, Melbourne, Australia
| | | | - Zeinab Abdullah
- Institute of Molecular Medicine and Experimental Immunology, University of Bonn, Bonn, Germany
| | - Robert P Finger
- Department of Ophthalmology, University of Bonn, Bonn, Germany; Department of Ophthalmology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
5
|
Peng J, Abdulla R, Liu X, He F, Xin X, Aisa HA. Polyphenol-Rich Extract of Apocynum venetum L. Leaves Protects Human Retinal Pigment Epithelial Cells against High Glucose-Induced Damage through Polyol Pathway and Autophagy. Nutrients 2024; 16:2944. [PMID: 39275261 PMCID: PMC11397065 DOI: 10.3390/nu16172944] [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: 07/07/2024] [Revised: 08/27/2024] [Accepted: 08/29/2024] [Indexed: 09/16/2024] Open
Abstract
Diabetic retinopathy (DR) is a specific microvascular problem of diabetes, which is mainly caused by hyperglycemia and may lead to rapid vision loss. Dietary polyphenols have been reported to decrease the risk of DR. Apocynum venetum L. leaves are rich in polyphenolic compounds and are popular worldwide for their health benefits as a national tea drink. Building on previous findings of antioxidant activity and aldose reductase inhibition of A. venetum, this study investigated the chemical composition of polyphenol-rich extract of A. venetum leaves (AVL) and its protective mechanism on ARPE-19 cells in hyperglycemia. Ninety-three compounds were identified from AVL by LC-MS/MS, including sixty-eight flavonoids, twenty-one organic acids, and four coumarins. AVL regulated the polyol pathway by decreasing the expression of aldose reductase and the content of sorbitol, enhancing the Na+K+-ATPase activity, and weakening intracellular oxidative stress effectively; it also could regulate the expression of autophagy-related proteins via the AMPK/mTOR/ULK1 signaling pathway to maintain intracellular homeostasis. AVL could restore the polyol pathway, inhibit oxidative stress, and maintain intracellular autophagy to protect cellular morphology and improve DR. The study reveals the phytochemical composition and protective mechanisms of AVL against DR, which could be developed as a functional food and/or candidate pharmaceutical, aiming for retina protection in diabetic retinopathy.
Collapse
Affiliation(s)
- Jun Peng
- The State Key Laboratory Basis Xinjiang Indigenous Medicinal Plant Resource, Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi 830011, China
- University of Chinese Academy of Sciences, Beijing 100039, China
| | - Rahima Abdulla
- The State Key Laboratory Basis Xinjiang Indigenous Medicinal Plant Resource, Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi 830011, China
| | - Xiaoyan Liu
- The State Key Laboratory Basis Xinjiang Indigenous Medicinal Plant Resource, Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi 830011, China
- University of Chinese Academy of Sciences, Beijing 100039, China
| | - Fei He
- The State Key Laboratory Basis Xinjiang Indigenous Medicinal Plant Resource, Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi 830011, China
| | - Xuelei Xin
- The State Key Laboratory Basis Xinjiang Indigenous Medicinal Plant Resource, Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi 830011, China
| | - Haji Akber Aisa
- The State Key Laboratory Basis Xinjiang Indigenous Medicinal Plant Resource, Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi 830011, China
| |
Collapse
|
6
|
Zhang Z, Liang F, Chang J, Shan X, Yin Z, Wang L, Li S. Autophagy in dry AMD: A promising therapeutic strategy for retinal pigment epithelial cell damage. Exp Eye Res 2024; 242:109889. [PMID: 38593971 DOI: 10.1016/j.exer.2024.109889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 04/04/2024] [Accepted: 04/05/2024] [Indexed: 04/11/2024]
Abstract
Dry age-related macular degeneration (AMD) is a prevalent clinical condition that leads to permanent damage to central vision and poses a significant threat to patients' visual health. Although the pathogenesis of dry AMD remains unclear, there is consensus on the role of retinal pigment epithelium (RPE) damage. Oxidative stress and chronic inflammation are major contributors to RPE cell damage, and the NOD-like receptor thermoprotein structural domain-associated protein 3 (NLRP3) inflammasome mediates the inflammatory response leading to apoptosis in RPE cells. Furthermore, lipofuscin accumulation results in oxidative stress, NLRP3 activation, and the development of vitelliform lesions, a hallmark of dry AMD, all of which may contribute to RPE dysfunction. The process of autophagy, involving the encapsulation, recognition, and transport of accumulated proteins and dead cells to the lysosome for degradation, is recognized as a significant pathway for cellular self-protection and homeostasis maintenance. Recently, RPE cell autophagy has been discovered to be closely linked to the development of macular degeneration, positioning autophagy as a cutting-edge research area in the realm of dry AMD. In this review, we present an overview of how lipofuscin, oxidative stress, and the NLRP3 inflammasome damage the RPE through their respective causal mechanisms. We summarized the connection between autophagy, oxidative stress, and NLRP3 inflammatory cytokines. Our findings suggest that targeting autophagy improves RPE function and sustains visual health, offering new perspectives for understanding the pathogenesis and clinical management of dry AMD.
Collapse
Affiliation(s)
- Zhao Zhang
- Tianjin University of Chinese Medicine, Tianjin, 300193, China; The First Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center of Traditional Chinese Medicine and Acupuncture and Moxibustion, Tianjin, 300193, China
| | - Fengming Liang
- The First Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center of Traditional Chinese Medicine and Acupuncture and Moxibustion, Tianjin, 300193, China.
| | - Jun Chang
- Tianjin University of Chinese Medicine, Tianjin, 300193, China; The First Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center of Traditional Chinese Medicine and Acupuncture and Moxibustion, Tianjin, 300193, China
| | - Xiaoqian Shan
- Tianjin University of Chinese Medicine, Tianjin, 300193, China; The First Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center of Traditional Chinese Medicine and Acupuncture and Moxibustion, Tianjin, 300193, China
| | - Zhixian Yin
- Hebei University of Technology, School of Electronics and Information Engineering, Tianjin, 300401, China
| | - Li Wang
- The First Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center of Traditional Chinese Medicine and Acupuncture and Moxibustion, Tianjin, 300193, China
| | - Shujiao Li
- Eye Hospital, China Academy of Chinese Medical Sciences, Beijing, 100040, China
| |
Collapse
|
7
|
Faura G, Studenovska H, Sekac D, Ellederova Z, Petrovski G, Eide L. The Effects of the Coating and Aging of Biodegradable Polylactic Acid Membranes on In Vitro Primary Human Retinal Pigment Epithelium Cells. Biomedicines 2024; 12:966. [PMID: 38790928 PMCID: PMC11117638 DOI: 10.3390/biomedicines12050966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 04/17/2024] [Accepted: 04/24/2024] [Indexed: 05/26/2024] Open
Abstract
Age-related macular degeneration (AMD) is the most frequent cause of blindness in developed countries. The replacement of dysfunctional human retinal pigment epithelium (hRPE) cells by the transplantation of in vitro-cultivated hRPE cells to the affected area emerges as a feasible strategy for regenerative therapy. Synthetic biomimetic membranes arise as powerful hRPE cell carriers, but as biodegradability is a requirement, it also poses a challenge due to its limited durability. hRPE cells exhibit several characteristics that putatively respond to the type of membrane carrier, and they can be used as biomarkers to evaluate and further optimize such membranes. Here, we analyze the pigmentation, transepithelial resistance, genome integrity, and maturation markers of hRPE cells plated on commercial polycarbonate (PC) versus in-house electrospun polylactide-based (PLA) membranes, both enabling separate apical/basolateral compartments. Our results show that PLA is superior to PC-based membranes for the cultivation of hRPEs, and the BEST1/RPE65 maturation markers emerge as the best biomarkers for addressing the quality of hRPE cultivated in vitro. The stability of the cultures was observed to be affected by PLA aging, which is an effect that could be partially palliated by the coating of the PLA membranes.
Collapse
Affiliation(s)
- Georgina Faura
- Department of Medical Biochemistry, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway
- CIDETEC, Basque Research and Technology Alliance (BRTA), 20014 Donostia-San Sebastián, Spain
| | - Hana Studenovska
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic, 162 00 Prague, Czech Republic;
| | - David Sekac
- Institute of Animal Physiology and Genetics, Academy of Sciences of the Czech Republic, 277 21 Libechov, Czech Republic; (D.S.); (Z.E.)
- Department of Cell Biology, Faculty of Science, Charles University, 128 00 Prague, Czech Republic
| | - Zdenka Ellederova
- Institute of Animal Physiology and Genetics, Academy of Sciences of the Czech Republic, 277 21 Libechov, Czech Republic; (D.S.); (Z.E.)
| | - Goran Petrovski
- Center for Eye Research and Innovative Diagnostics, Department of Ophthalmology, Oslo University Hospital and Institute for Clinical Medicine, University of Oslo, 0424 Oslo, Norway;
- Norwegian Center for Stem Cell Research, Oslo University Hospital, 0424 Oslo, Norway
- Department of Ophthalmology, University Hospital Centre, University of Split School of Medicine, 21000 Split, Croatia
- UKLO Network, University St. Kliment Ohridski, 7000 Bitola, North Macedonia
| | - Lars Eide
- Department of Medical Biochemistry, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway
- Department of Medical Biochemistry, Oslo University Hospital, 0424 Oslo, Norway
| |
Collapse
|
8
|
Schwartz RE, Conboy IM. Non-Intrinsic, Systemic Mechanisms of Cellular Senescence. Cells 2023; 12:2769. [PMID: 38132089 PMCID: PMC10741531 DOI: 10.3390/cells12242769] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 11/30/2023] [Accepted: 12/01/2023] [Indexed: 12/23/2023] Open
Abstract
Cellular senescence is believed to contribute to aging and disease through the activity of secreted factors that promote inflammation, remodel the extracellular matrix, and adversely modify the behavior of non-senescent cells. While the markers and properties of senescent cells are still under investigation, it is postulated that cellular senescence manifests in vivo as the consequence of cellular damage that accumulates and becomes exacerbated with time. Yet, the notions that senescence has a solely intrinsic and time-dependent nature are questioned by the rapid induction of senescence in young mice and young cells in vitro by exposure to blood from aged animals. Here, we review some of the research on the systemically present factors that increase with age and may contribute to extrinsically induced senescence or "bystander senescence". These include proteins, reactive oxygen species, lipids, and nucleic acids, which may be present in individual soluble form, in vesicles, and in non-membranous multi-component macromolecules.
Collapse
Affiliation(s)
| | - Irina M. Conboy
- Department of Bioengineering, University of California Berkeley, Berkeley, CA 94720, USA;
| |
Collapse
|
9
|
Weinberg J, Gaur M, Swaroop A, Taylor A. Proteostasis in aging-associated ocular disease. Mol Aspects Med 2022; 88:101157. [PMID: 36459837 PMCID: PMC9742340 DOI: 10.1016/j.mam.2022.101157] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 11/15/2022] [Indexed: 11/30/2022]
Abstract
Vision impairment has devastating consequences for the quality of human life. The cells and tissues associated with the visual process must function throughout one's life span and maintain homeostasis despite exposure to a variety of insults. Maintenance of the proteome is termed proteostasis, and is vital for normal cellular functions, especially at an advanced age. Here we describe basic aspects of proteostasis, from protein synthesis and folding to degradation, and discuss the current status of the field with a particular focus on major age-related eye diseases: age-related macular degeneration, cataract, and glaucoma. Our intent is to allow vision scientists to determine where and how to harness the proteostatic machinery for extending functional homeostasis in the aging retina, lens, and trabecular meshwork. Several common themes have emerged despite these tissues having vastly different metabolisms. Continued exposure to insults, including chronic stress with advancing age, increases proteostatic burden and reduces the fidelity of the degradation machineries including the ubiquitin-proteasome and the autophagy-lysosome systems that recognize and remove damaged proteins. This "double jeopardy" results in an exponential accumulation of cytotoxic proteins with advancing age. We conclude with a discussion of the challenges in maintaining an appropriate balance of protein synthesis and degradation pathways, and suggest that harnessing proteostatic capacities should provide new opportunities to design interventions for attenuating age-related eye diseases before they limit sight.
Collapse
Affiliation(s)
- Jasper Weinberg
- Laboratory for Nutrition and Vision Research, USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, 02111, USA
| | - Mohita Gaur
- Neurobiology, Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Anand Swaroop
- Neurobiology, Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Allen Taylor
- Laboratory for Nutrition and Vision Research, USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, 02111, USA.
| |
Collapse
|
10
|
Fehér J, Élő Á, István L, Nagy ZZ, Radák Z, Scuderi G, Artico M, Kovács I. Microbiota mitochondria disorders as hubs for early age-related macular degeneration. GeroScience 2022; 44:2623-2653. [PMID: 35978068 PMCID: PMC9385247 DOI: 10.1007/s11357-022-00620-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 07/01/2022] [Indexed: 01/07/2023] Open
Abstract
Age-related macular degeneration (AMD) is a progressive neurodegenerative disease affecting the central area (macula lutea) of the retina. Research on the pathogenic mechanism of AMD showed complex cellular contribution governed by such risk factors as aging, genetic predisposition, diet, and lifestyle. Recent studies suggested that microbiota is a transducer and a modifier of risk factors for neurodegenerative diseases, and mitochondria may be one of the intracellular targets of microbial signaling molecules. This review explores studies supporting a new concept on the contribution of microbiota-mitochondria disorders to AMD. We discuss metabolic, vascular, immune, and neuronal mechanism in AMD as well as key alterations of photoreceptor cells, retinal pigment epithelium (RPE), Bruch's membrane, choriocapillaris endothelial, immune, and neuronal cells. Special attention was paid to alterations of mitochondria contact sites (MCSs), an organelle network of mitochondria, endoplasmic reticulum, lipid droplets (LDs), and peroxisomes being documented based on our own electron microscopic findings from surgically removed human eyes. Morphometry of Bruch's membrane lipids and proteoglycans has also been performed in early AMD and aged controls. Microbial metabolites (short-chain fatty acids, polyphenols, and secondary bile acids) and microbial compounds (lipopolysaccharide, peptidoglycan, and bacterial DNA)-now called postbiotics-in addition to local effects on resident microbiota and mucous membrane, regulate systemic metabolic, vascular, immune, and neuronal mechanisms in normal conditions and in various common diseases. We also discuss their antioxidant, anti-inflammatory, and metabolic effects as well as experimental and clinical observations on regulating the main processes of photoreceptor renewal, mitophagy, and autophagy in early AMD. These findings support an emerging concept that microbiota-mitochondria disorders may be a crucial pathogenic mechanism of early AMD; and similarly, to other age-related neurodegenerative diseases, new treatment approaches should be targeted at these disorders.
Collapse
Affiliation(s)
- János Fehér
- PRIMAVERA Program, Nutripharma Hungaria Ltd., Budapest, Hungary
| | - Ágnes Élő
- grid.11804.3c0000 0001 0942 9821Department of Ophthalmology, Semmelweis University, Budapest, Hungary
| | - Lilla István
- grid.11804.3c0000 0001 0942 9821Department of Ophthalmology, Semmelweis University, Budapest, Hungary
| | - Zoltán Zsolt Nagy
- grid.11804.3c0000 0001 0942 9821Department of Ophthalmology, Semmelweis University, Budapest, Hungary
| | - Zsolt Radák
- grid.472475.70000 0000 9243 1481Research Institute of Sport Science, University of Physical Education, Budapest, Hungary
| | - Gianluca Scuderi
- grid.7841.aOphthalmology Unit, NESMOS Department, Sant’Andrea Hospital, Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy
| | - Marco Artico
- grid.417007.5Department of Sensory Organs, “Sapienza” University of Rome, Roma, Italy
| | - Illés Kovács
- grid.11804.3c0000 0001 0942 9821Department of Ophthalmology, Semmelweis University, Budapest, Hungary ,grid.5386.8000000041936877XDepartment of Ophthalmology, Weill Cornell Medical College, New York City, NY USA
| |
Collapse
|
11
|
Subretinal Implantation of Human Primary RPE Cells Cultured on Nanofibrous Membranes in Minipigs. Biomedicines 2022; 10:biomedicines10030669. [PMID: 35327471 PMCID: PMC8945676 DOI: 10.3390/biomedicines10030669] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 02/19/2022] [Accepted: 02/22/2022] [Indexed: 11/28/2022] Open
Abstract
Purpose: The development of primary human retinal pigmented epithelium (hRPE) for clinical transplantation purposes on biodegradable scaffolds is indispensable. We hereby report the results of the subretinal implantation of hRPE cells on nanofibrous membranes in minipigs. Methods: The hRPEs were collected from human cadaver donor eyes and cultivated on ultrathin nanofibrous carriers prepared via the electrospinning of poly(L-lactide-co-DL-lactide) (PDLLA). “Libechov” minipigs (12–36 months old) were used in the study, supported by preoperative tacrolimus immunosuppressive therapy. The subretinal implantation of the hRPE-nanofibrous carrier was conducted using general anesthesia via a custom-made injector during standard three-port 23-gauge vitrectomy, followed by silicone oil endotamponade. The observational period lasted 1, 2, 6 and 8 weeks, and included in vivo optical coherence tomography (OCT) of the retina, as well as post mortem immunohistochemistry using the following antibodies: HNAA and STEM121 (human cell markers); Bestrophin and CRALBP (hRPE cell markers); peanut agglutining (PNA) (cone photoreceptor marker); PKCα (rod bipolar marker); Vimentin, GFAP (macroglial markers); and Iba1 (microglial marker). Results: The hRPEs assumed cobblestone morphology, persistent pigmentation and measurable trans-epithelial electrical resistance on the nanofibrous PDLLA carrier. The surgical delivery of the implants in the subretinal space of the immunosuppressed minipigs was successfully achieved and monitored by fundus imaging and OCT. The implanted hRPEs were positive for HNAA and STEM121 and were located between the minipig’s neuroretina and RPE layers at week 2 post-implantation, which was gradually attenuated until week 8. The neuroretina over the implants showed rosette or hypertrophic reaction at week 6. The implanted cells expressed the typical RPE marker bestrophin throughout the whole observation period, and a gradual diminishing of the CRALBP expression in the area of implantation at week 8 post-implantation was observed. The transplanted hRPEs appeared not to form a confluent layer and were less capable of keeping the inner and outer retinal segments intact. The cone photoreceptors adjacent to the implant scaffold were unchanged initially, but underwent a gradual change in structure after hRPE implantation; the retina above and below the implant appeared relatively healthy. The glial reaction of the transplanted and host retina showed Vimentin and GFAP positivity from week 1 onward. Microglial activation appeared in the retinal area of the transplant early after the surgery, which seemed to move into the transplant area over time. Conclusions: The differentiated hRPEs can serve as an alternative cell source for RPE replacement in animal studies. These cells can be cultivated on nanofibrous PDLLA and implanted subretinally into minipigs using standard 23-gauge vitrectomy and implantation injector. The hRPE-laden scaffolds demonstrated relatively good incorporation into the host retina over an eight-week observation period, with some indication of a gliotic scar formation, and a likely neuroinflammatory response in the transplanted area despite the use of immunosuppression.
Collapse
|
12
|
Zibetti C. Deciphering the Retinal Epigenome during Development, Disease and Reprogramming: Advancements, Challenges and Perspectives. Cells 2022; 11:cells11050806. [PMID: 35269428 PMCID: PMC8908986 DOI: 10.3390/cells11050806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/15/2022] [Accepted: 02/18/2022] [Indexed: 02/01/2023] Open
Abstract
Retinal neurogenesis is driven by concerted actions of transcription factors, some of which are expressed in a continuum and across several cell subtypes throughout development. While seemingly redundant, many factors diversify their regulatory outcome on gene expression, by coordinating variations in chromatin landscapes to drive divergent retinal specification programs. Recent studies have furthered the understanding of the epigenetic contribution to the progression of age-related macular degeneration, a leading cause of blindness in the elderly. The knowledge of the epigenomic mechanisms that control the acquisition and stabilization of retinal cell fates and are evoked upon damage, holds the potential for the treatment of retinal degeneration. Herein, this review presents the state-of-the-art approaches to investigate the retinal epigenome during development, disease, and reprogramming. A pipeline is then reviewed to functionally interrogate the epigenetic and transcriptional networks underlying cell fate specification, relying on a truly unbiased screening of open chromatin states. The related work proposes an inferential model to identify gene regulatory networks, features the first footprinting analysis and the first tentative, systematic query of candidate pioneer factors in the retina ever conducted in any model organism, leading to the identification of previously uncharacterized master regulators of retinal cell identity, such as the nuclear factor I, NFI. This pipeline is virtually applicable to the study of genetic programs and candidate pioneer factors in any developmental context. Finally, challenges and limitations intrinsic to the current next-generation sequencing techniques are discussed, as well as recent advances in super-resolution imaging, enabling spatio-temporal resolution of the genome.
Collapse
Affiliation(s)
- Cristina Zibetti
- Department of Ophthalmology, Institute of Clinical Medicine, University of Oslo, Kirkeveien 166, Building 36, 0455 Oslo, Norway
| |
Collapse
|
13
|
Chang KC, Liu PF, Chang CH, Lin YC, Chen YJ, Shu CW. The interplay of autophagy and oxidative stress in the pathogenesis and therapy of retinal degenerative diseases. Cell Biosci 2022; 12:1. [PMID: 34980273 PMCID: PMC8725349 DOI: 10.1186/s13578-021-00736-9] [Citation(s) in RCA: 90] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 12/19/2021] [Indexed: 12/27/2022] Open
Abstract
Oxidative stress is mainly caused by intracellular reactive oxygen species (ROS) production, which is highly associated with normal physiological homeostasis and the pathogenesis of diseases, particularly ocular diseases. Autophagy is a self-clearance pathway that removes oxidized cellular components and regulates cellular ROS levels. ROS can modulate autophagy activity through transcriptional and posttranslational mechanisms. Autophagy further triggers transcription factor activation and degrades impaired organelles and proteins to eliminate excessive ROS in cells. Thus, autophagy may play an antioxidant role in protecting ocular cells from oxidative stress. Nevertheless, excessive autophagy may cause autophagic cell death. In this review, we summarize the mechanisms of interaction between ROS and autophagy and their roles in the pathogenesis of several ocular diseases, including glaucoma, age-related macular degeneration (AMD), diabetic retinopathy (DR), and optic nerve atrophy, which are major causes of blindness. The autophagy modulators used to treat ocular diseases are further discussed. The findings of the studies reviewed here might shed light on the development and use of autophagy modulators for the future treatment of ocular diseases.
Collapse
Affiliation(s)
- Kun-Che Chang
- Department of Ophthalmology and Neurobiology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Pei-Feng Liu
- Department of Biomedical Science and Environmental Biology, PhD Program in Life Science, College of Life Science, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,Center for Cancer Research, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chia-Hsuan Chang
- Institute of BioPharmaceutical Sciences, National Sun Yat-Sen University, No. 70, Lianhai Rd., Gushan Dist., Kaohsiung, 80424, Taiwan
| | - Ying-Cheng Lin
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Yen-Ju Chen
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan.,Division of Allergy, Immunology and Rheumatology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Chih-Wen Shu
- Institute of BioPharmaceutical Sciences, National Sun Yat-Sen University, No. 70, Lianhai Rd., Gushan Dist., Kaohsiung, 80424, Taiwan.
| |
Collapse
|
14
|
Asatryan A, Calandria JM, Kautzmann MAI, Jun B, Gordon WC, Do KV, Bhattacharjee S, Pham TL, Bermúdez V, Mateos MV, Heap J, Bazan NG. New Retinal Pigment Epithelial Cell Model to Unravel Neuroprotection Sensors of Neurodegeneration in Retinal Disease. Front Neurosci 2022; 16:926629. [PMID: 35873810 PMCID: PMC9301569 DOI: 10.3389/fnins.2022.926629] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 05/30/2022] [Indexed: 01/02/2023] Open
Abstract
Retinal pigment epithelial (RPE) cells sustain photoreceptor integrity, and when this function is disrupted, retinal degenerations ensue. Herein, we characterize a new cell line from human RPE that we termed ABC. These cells remarkably recapitulate human eye native cells. Distinctive from other epithelia, RPE cells originate from the neural crest and follow a neural development but are terminally differentiated into "epithelial" type, thus sharing characteristics with their neuronal lineages counterparts. Additionally, they form microvilli, tight junctions, and honeycomb packing and express distinctive markers. In these cells, outer segment phagocytosis, phagolysosome fate, phospholipid metabolism, and lipid mediator release can be studied. ABC cells display higher resistance to oxidative stress and are protected from senescence through mTOR inhibition, making them more stable in culture. The cells are responsive to Neuroprotectin D1 (NPD1), which downregulates inflammasomes and upregulates antioxidant and anti-inflammatory genes. ABC gene expression profile displays close proximity to native RPE lineage, making them a reliable cell system to unravel signaling in uncompensated oxidative stress (UOS) and retinal degenerative disease to define neuroprotection sites.
Collapse
Affiliation(s)
- Aram Asatryan
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA, United States
| | - Jorgelina M Calandria
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA, United States
| | - Marie-Audrey I Kautzmann
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA, United States
| | - Bokkyoo Jun
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA, United States
| | - William C Gordon
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA, United States
| | - Khanh V Do
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA, United States
| | - Surjyadipta Bhattacharjee
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA, United States
| | - Thang L Pham
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA, United States
| | - Vicente Bermúdez
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA, United States
| | - Melina Valeria Mateos
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA, United States
| | - Jessica Heap
- 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
| |
Collapse
|
15
|
Amyloid-Beta Peptides and Activated Astroglia Impairs Proliferation of Nerve Growth Factor Releasing Cells In Vitro: Implication for Encapsulated Cell Biodelivery-Mediated AD Therapy. Cells 2021; 10:cells10112834. [PMID: 34831056 PMCID: PMC8616486 DOI: 10.3390/cells10112834] [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: 08/17/2021] [Revised: 09/29/2021] [Accepted: 10/14/2021] [Indexed: 01/13/2023] Open
Abstract
Alzheimer’s disease (AD) treatment is constrained due to the inability of peripherally administered therapeutic molecules to cross the blood–brain barrier. Encapsulated cell biodelivery (ECB) devices, a tissue-targeted approach for local drug release, was previously optimized for human mature nerve growth factor (hmNGF) delivery in AD patients but was found to have reduced hmNGF release over time. To understand the reason behind reduced ECB efficacy, we exposed hmNGF-releasing cells (NGC0211) in vitro to human cerebrospinal fluid (CSF) obtained from Subjective Cognitive Impairment (SCI), Lewy Body Dementia (LBD), and AD patients. Subsequently, we exposed NGC0211 cells directly to AD-related factors like amyloid-β peptides (Aβ40/42) or activated astrocyte-conditioned medium (Aβ40/42/IL-1β/TNFα-treated) and evaluated biochemical stress markers, cell death indicators, cell proliferation marker (Ki67), and hmNGF release. We found that all patients’ CSF significantly reduced hmNGF release from NGC0211 cells in vitro. Aβ40/42, inflammatory molecules, and activated astrocytes significantly affected NGC0211 cell proliferation without altering hmNGF release or other parameters important for essential functions of the NGC0211 cells. Long-term constant cell proliferation within the ECB device is critically important to maintain a steady cell population needed for stable mNGF release. These data show hampered proliferation of NGC0211 cells, which may lead to a decline of the NGC0211 cell population in ECBs, thereby reducing hmNGF release. Our study highlights the need for future studies to strengthen ECB-mediated long-term drug delivery approaches.
Collapse
|
16
|
Targeting Lysosomes to Reverse Hydroquinone-Induced Autophagy Defects and Oxidative Damage in Human Retinal Pigment Epithelial Cells. Int J Mol Sci 2021; 22:ijms22169042. [PMID: 34445748 PMCID: PMC8396439 DOI: 10.3390/ijms22169042] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/19/2021] [Accepted: 08/19/2021] [Indexed: 01/09/2023] Open
Abstract
In age-related macular degeneration (AMD), hydroquinone (HQ)-induced oxidative damage in retinal pigment epithelium (RPE) is believed to be an early event contributing to dysregulation of inflammatory cytokines and vascular endothelial growth factor (VEGF) homeostasis. However, the roles of antioxidant mechanisms, such as autophagy and the ubiquitin-proteasome system, in modulating HQ-induced oxidative damage in RPE is not well-understood. This study utilized an in-vitro AMD model involving the incubation of human RPE cells (ARPE-19) with HQ. In comparison to hydrogen peroxide (H2O2), HQ induced fewer reactive oxygen species (ROS) but more oxidative damage as characterized by protein carbonyl levels, mitochondrial dysfunction, and the loss of cell viability. HQ blocked the autophagy flux and increased proteasome activity, whereas H2O2 did the opposite. Moreover, the lysosomal membrane-stabilizing protein LAMP2 and cathepsin D levels declined with HQ exposure, suggesting loss of lysosomal membrane integrity and function. Accordingly, HQ induced lysosomal alkalization, thereby compromising the acidic pH needed for optimal lysosomal degradation. Pretreatment with MG132, a proteasome inhibitor and lysosomal stabilizer, upregulated LAMP2 and autophagy and prevented HQ-induced oxidative damage in wildtype RPE cells but not cells transfected with shRNA against ATG5. This study demonstrated that lysosomal dysfunction underlies autophagy defects and oxidative damage induced by HQ in human RPE cells and supports lysosomal stabilization with the proteasome inhibitor MG132 as a potential remedy for oxidative damage in RPE and AMD.
Collapse
|
17
|
Dhingra A, Sharp RC, Kim T, Popov AV, Ying GS, Pietrofesa RA, Park K, Christofidou-Solomidou M, Boesze-Battaglia K. Assessment of a Small Molecule Synthetic Lignan in Enhancing Oxidative Balance and Decreasing Lipid Accumulation in Human Retinal Pigment Epithelia. Int J Mol Sci 2021; 22:5764. [PMID: 34071220 PMCID: PMC8198017 DOI: 10.3390/ijms22115764] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 05/24/2021] [Accepted: 05/26/2021] [Indexed: 01/03/2023] Open
Abstract
Visual function depends on the intimate structural, functional and metabolic interactions between the retinal pigment epithelium (RPE) and the neural retina. The daily phagocytosis of the photoreceptor outer segment tips by the overlaying RPE provides essential nutrients for the RPE itself and photoreceptors through intricate metabolic synergy. Age-related retinal changes are often characterized by metabolic dysregulation contributing to increased lipid accumulation and peroxidation as well as the release of proinflammatory cytokines. LGM2605 is a synthetic lignan secoisolariciresinol diglucoside (SDG) with free radical scavenging, antioxidant and anti-inflammatory properties demonstrated in diverse in vitro and in vivo inflammatory disease models. In these studies, we tested the hypothesis that LGM2605 may be an attractive small-scale therapeutic that protects RPE against inflammation and restores its metabolic capacity under lipid overload. Using an in vitro model in which loss of the autophagy protein, LC3B, results in defective phagosome degradation and metabolic dysregulation, we show that lipid overload results in increased gasdermin cleavage, IL-1 β release, lipid accumulation and decreased oxidative capacity. The addition of LGM2605 resulted in enhanced mitochondrial capacity, decreased lipid accumulation and amelioration of IL-1 β release in a model of defective lipid homeostasis. Collectively, these studies suggest that lipid overload decreases mitochondrial function and increases the inflammatory response, with LGM2605 acting as a protective agent.
Collapse
Affiliation(s)
- Anuradha Dhingra
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (A.D.); (R.C.S.)
| | - Rachel C. Sharp
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (A.D.); (R.C.S.)
| | - Taewan Kim
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Anatoliy V. Popov
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Gui-Shuang Ying
- Center for Preventive Ophthalmology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (G.-S.Y.); (K.P.)
| | - Ralph A. Pietrofesa
- Department of Medicine, School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (R.A.P.); (M.C.-S.)
| | - Kyewon Park
- Center for Preventive Ophthalmology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (G.-S.Y.); (K.P.)
| | - Melpo Christofidou-Solomidou
- Department of Medicine, School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (R.A.P.); (M.C.-S.)
| | - Kathleen Boesze-Battaglia
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (A.D.); (R.C.S.)
| |
Collapse
|
18
|
The Role of Autophagy in Eye Diseases. Life (Basel) 2021; 11:life11030189. [PMID: 33673657 PMCID: PMC7997177 DOI: 10.3390/life11030189] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 02/23/2021] [Accepted: 02/24/2021] [Indexed: 12/18/2022] Open
Abstract
Autophagy is a catabolic process that ensures homeostasis in the cells of our organism. It plays a crucial role in protecting eye cells against oxidative damage and external stress factors. Ocular pathologies of high incidence, such as age-related macular degeneration, cataracts, glaucoma, and diabetic retinopathy are of multifactorial origin and are associated with genetic, environmental factors, age, and oxidative stress, among others; the latter factor is one of the most influential in ocular diseases, directly affecting the processes of autophagy activity. Alteration of the normal functioning of autophagy processes can interrupt organelle turnover, leading to the accumulation of cellular debris and causing physiological dysfunction of the eye. The aim of this study is to review research on the role of autophagy processes in the main ocular pathologies, which have a high incidence and result in high costs for the health system. Considering the role of autophagy processes in cell homeostasis and cell viability, the control and modulation of autophagy processes in ocular pathologies could constitute a new therapeutic approach.
Collapse
|
19
|
Feng J, Tan W, Li T, Yan Q, Zhu H, Sun X. Human retinal pigment epithelial cells are protected against hypoxia by BNIP3. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1502. [PMID: 33313247 PMCID: PMC7729317 DOI: 10.21037/atm-20-7145] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Background Hypoxia has been implicated in the process of retinal pigment epithelium (RPE) dysfunction. However, recent studies suggest that hypoxia contributes to survival rather than cell death through induction of Bcl-2/adenovirus E1B 19-kDa interacting protein 3 (BNIP3)-dependent autophagy. In contrast, persistent oxidative stress was found to result in autophagy dysregulation in RPE cells. These seemingly contradictory findings led us to investigate the potential role of BNIP3, a crucial mediator of hypoxia-induced autophagy, in the context of hypoxic RPE cells. Methods Human RPE D407 cells were treated with low-oxygen conditions, and cell growth, apoptosis, and autophagy was assessed by Cell Counting Kit-8 assay, flow cytometry analysis and immunofluorescence staining, respectively. Results Hypoxic conditions simultaneously triggered a large amount of apoptosis and inhibited autophagy. Moreover, hypoxia led to severe impairments, including the stimulation of reactive oxygen species, and reduction of mitochondrial membrane potential, and adenosine triphosphate production. The stimulation of autophagy by rapamycin inhibited hypoxia-induced severe impairments to a great extent. Interestingly, similar results were observed for BNIP3 overexpression, which can be largely blocked by 3-MA, a well-defined inhibitor of autophagy. Moreover, BNIP3 knockdown further aggravated hypoxia-induced impairments in D407 cells, which can be reversed by rapamycin. Conclusions Collectively, these results indicated that BNIP3 can protect human retinal pigmented epithelial cells under hypoxic conditions by inducing autophagy.
Collapse
Affiliation(s)
- Jingyang Feng
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University, School of Medicine, Shanghai, China.,Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China
| | - Wei Tan
- Department of Ophthalmology, Zunyi First People's Hospital, The Third Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Tong Li
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University, School of Medicine, Shanghai, China.,Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China
| | - Quan Yan
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University, School of Medicine, Shanghai, China.,Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China
| | - Hong Zhu
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University, School of Medicine, Shanghai, China.,Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China.,Shanghai Key Laboratory of Fundus Diseases, Shanghai, China
| | - Xiaodong Sun
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University, School of Medicine, Shanghai, China.,Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China.,Shanghai Key Laboratory of Fundus Diseases, Shanghai, China
| |
Collapse
|
20
|
Kaarniranta K, Uusitalo H, Blasiak J, Felszeghy S, Kannan R, Kauppinen A, Salminen A, Sinha D, Ferrington D. Mechanisms of mitochondrial dysfunction and their impact on age-related macular degeneration. Prog Retin Eye Res 2020; 79:100858. [PMID: 32298788 PMCID: PMC7650008 DOI: 10.1016/j.preteyeres.2020.100858] [Citation(s) in RCA: 319] [Impact Index Per Article: 63.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 03/18/2020] [Accepted: 03/19/2020] [Indexed: 12/21/2022]
Abstract
Oxidative stress-induced damage to the retinal pigment epithelium (RPE) is considered to be a key factor in age-related macular degeneration (AMD) pathology. RPE cells are constantly exposed to oxidative stress that may lead to the accumulation of damaged cellular proteins, lipids, nucleic acids, and cellular organelles, including mitochondria. The ubiquitin-proteasome and the lysosomal/autophagy pathways are the two major proteolytic systems to remove damaged proteins and organelles. There is increasing evidence that proteostasis is disturbed in RPE as evidenced by lysosomal lipofuscin and extracellular drusen accumulation in AMD. Nuclear factor-erythroid 2-related factor-2 (NFE2L2) and peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α) are master transcription factors in the regulation of antioxidant enzymes, clearance systems, and biogenesis of mitochondria. The precise cause of RPE degeneration and the onset and progression of AMD are not fully understood. However, mitochondria dysfunction, increased reactive oxygen species (ROS) production, and mitochondrial DNA (mtDNA) damage are observed together with increased protein aggregation and inflammation in AMD. In contrast, functional mitochondria prevent RPE cells damage and suppress inflammation. Here, we will discuss the role of mitochondria in RPE degeneration and AMD pathology focused on mtDNA damage and repair, autophagy/mitophagy signaling, and regulation of inflammation. Mitochondria are putative therapeutic targets to prevent or treat AMD.
Collapse
Affiliation(s)
- Kai Kaarniranta
- Department of Ophthalmology, Institute of Clinical Medicine, University of Eastern Finland and Kuopio University Hospital, P.O. Box 1627, FI-70211, Kuopio, Finland.
| | - Hannu Uusitalo
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland and Tays Eye Centre, Tampere University Hospital, P.O.Box 2000, 33521 Tampere, Finland
| | - Janusz Blasiak
- Department of Molecular Genetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236, Lodz, Poland
| | - Szabolcs Felszeghy
- Department of Biomedicine, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Ram Kannan
- The Stephen J. Ryan Initiative for Macular Research (RIMR), Doheny Eye Institute, 1355 San Pablo St, Los Angeles, CA, 90033, USA
| | - Anu Kauppinen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Antero Salminen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Debasish Sinha
- Glia Research Laboratory, Department of Ophthalmology, University of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, PA 15224, USA; Wilmer Eye Institute, The Johns Hopkins University School of Medicine, Room M035 Robert and Clarice Smith Bldg, 400 N Broadway, Baltimore, MD, 21287, USA
| | - Deborah Ferrington
- Department of Ophthalmology and Visual Neurosciences, 2001 6th St SE, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
21
|
Nita M, Grzybowski A. Interplay between reactive oxygen species and autophagy in the course of age-related macular degeneration. EXCLI JOURNAL 2020; 19:1353-1371. [PMID: 33192217 PMCID: PMC7658465 DOI: 10.17179/excli2020-2915] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 09/18/2020] [Indexed: 12/11/2022]
Abstract
Pathological biomolecules such as lipofuscin, methylglyoxal-modified proteins (the major precursors of advanced glycationend products), misfolding protein deposits and dysfunctional mitochondria are source of oxidative stress and act as strong autophagic stimulators in age-related macular degeneration. Disturbed autophagy accelerates progression of the disease, since it leads to retinal cells' death and activates inflammation by the interplay with the NLRP3 inflammasome complex. Vascular dysfunction and hypoxia, as well as circulating autoantibodies against autophagy regulators (anti-S100A9, anti-ANXA5, and anti-HSPA8, A9 and B4) compromise an autophagy-mediated mechanism as well. Metformin, the autophagic stimulator, may act as a senostatic drug to inhibit the senescent phenotype in the age-related macular degeneration. PGC-1α , Sirt1 and AMPK represent new therapeutic targets for interventions in this disease.
Collapse
Affiliation(s)
- Malgorzata Nita
- Domestic and Specialized Medicine Centre "Dilmed" Katowice, Poland
| | - Andrzej Grzybowski
- Department of Ophthalmolgy, Medical Faculty, University of Warmia and Mazury, Olsztyn, Poland.,Institute for Research in Ophthalmology, Poznań, Poland
| |
Collapse
|
22
|
Tan W, Zou J, Yoshida S, Jiang B, Zhou Y. The Role of Inflammation in Age-Related Macular Degeneration. Int J Biol Sci 2020; 16:2989-3001. [PMID: 33061811 PMCID: PMC7545698 DOI: 10.7150/ijbs.49890] [Citation(s) in RCA: 154] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 09/10/2020] [Indexed: 02/06/2023] Open
Abstract
Age-related macular degeneration (AMD) is a blinding eye disease which incidence gradually increases with age. Inflammation participates in AMD pathogenesis, including choroidal neovascularization and geographic atrophy. It is also a kind of self-protective regulation from injury for the eyes. In this review, we described inflammation in AMD pathogenesis, summarized the roles played by inflammation-related cytokines, including pro-inflammatory and anti-inflammatory cytokines, as well as leukocytes (macrophages, dendritic cells, neutrophils, T lymphocytes and B lymphocytes) in the innate or adaptive immunity in AMD. Possible clinical applications such as potential diagnostic biomarkers and anti-inflammatory therapies were also discussed. This review overviews the inflammation as a target of novel effective therapies in treating AMD.
Collapse
Affiliation(s)
- Wei Tan
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China.,Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan 410011, China
| | - Jingling Zou
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China.,Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan 410011, China
| | - Shigeo Yoshida
- Department of Ophthalmology, Kurume University School of Medicine, Kurume, Fukuoka 830-0011, Japan
| | - Bing Jiang
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China.,Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan 410011, China
| | - Yedi Zhou
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China.,Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan 410011, China
| |
Collapse
|
23
|
Jabbehdari S, Handa JT. Oxidative stress as a therapeutic target for the prevention and treatment of early age-related macular degeneration. Surv Ophthalmol 2020; 66:423-440. [PMID: 32961209 DOI: 10.1016/j.survophthal.2020.09.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 09/10/2020] [Accepted: 09/14/2020] [Indexed: 12/13/2022]
Abstract
Age-related macular degeneration, the leading cause of irreversible visual loss among older adults in developed countries, is a chronic, multifactorial, and progressive disease with the development of painless, central vision loss. Retinal pigment epithelial cell dysfunction is a core change in age-related macular degeneration that results from aging and the accumulated effects of genetic and environmental factors that, in part, is both caused by and leads to oxidative stress. In this review, we describe the role of oxidative stress, the cytoprotective oxidative stress pathways, and the impact of oxidative stress on critical cellular processes involved in age-related macular degeneration pathobiology. We also offer targeted therapy that may define how antioxidant therapy can either prevent or improve specific stages of age-related macular degeneration.
Collapse
Affiliation(s)
- Sayena Jabbehdari
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, Illinois, USA
| | - James T Handa
- Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, Maryland, USA.
| |
Collapse
|
24
|
Thermogenic Activation Downregulates High Mitophagy Rate in Human Masked and Mature Beige Adipocytes. Int J Mol Sci 2020; 21:ijms21186640. [PMID: 32927882 PMCID: PMC7555361 DOI: 10.3390/ijms21186640] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 09/07/2020] [Accepted: 09/07/2020] [Indexed: 12/16/2022] Open
Abstract
Thermogenic brown and beige adipocytes oxidize metabolic substrates producing heat, mainly by the mitochondrial uncoupling protein UCP1, and can thus counteract obesity. Masked beige adipocytes possess white adipocyte-like morphology, but can be made thermogenic by adrenergic stimuli. We investigated the regulation of mitophagy upon thermogenic activation of human masked and mature beige adipocytes. Human primary abdominal subcutaneous adipose-derived stromal cells (hASCs) and Simpson-Golabi-Behmel syndrome (SGBS) preadipocytes were differentiated to white and beige adipocytes, then their cAMP-induced thermogenic potential was assessed by detecting increased expressions of UCP1, mitochondrial DNA content and respiratory chain complex subunits. cAMP increased the thermogenic potential of white adipocytes similarly to beige ones, indicating the presence of a masked beige population. In unstimulated conditions, a high autophagic flux and mitophagy rates (demonstrated by LC3 punctae and TOM20 co-immunostaining) were observed in white adipocytes, while these were lower in beige adipocytes. Silencing and gene expression experiments showed that the ongoing mitophagy was Parkin-independent. cAMP treatment led to the downregulation of mitophagy through PKA in both types of adipocytes, resulting in more fragmented mitochondria and increased UCP1 levels. Our data indicates that mitophagy is repressed upon encountering a short-term adrenergic stimulus, as a fast regulatory mechanism to provide high mitochondrial content for thermogenesis.
Collapse
|
25
|
Kubicka-Trząska A, Karska-Basta I, Żuber-Łaskawiec K. Autophagy: A new insight into pathogenesis and treatment possibilities in age-related macular degeneration. POSTEP HIG MED DOSW 2020. [DOI: 10.5604/01.3001.0014.2495] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Age-related macular degeneration (AMD) is a significant problem in healthcare, because it is a leading cause of central vision loss in individuals over 50 years old in well-developed countries. Pathogenesis of AMD is multifactorial and still not completely understood. Proven risk factors include the following: natural senescence of retina, oxidative stress, complement activation, chronic subretinal inflammatory reaction, genetic and environmental factors. Data on links between autophagy and AMD development are being raised. Autophagy is a cellular
process involving the degradation of long-lived proteins and damaged fragments and components
of cells; it is responsible for the maintenance of dynamic intracellular homeostasis
and it enables cell survival under stress conditions. Disturbances of autophagy mechanisms,
i.e. its activation or inhibition, may lead to the development of many various pathologies.
Thus, autophagy plays a dual role, as a mechanism responsible for protecting or killing cells.
The paper describes autophagy mechanisms and their role in the natural process of retinal cells
senescence and presents the autophagy impairment as a crucial cause of AMD development.
We also describe the impact of intravitreal anti-VEGF therapy on retinal autophagy mechanisms
and potential new therapeutic modalities for AMD based on autophagy modulation.
Collapse
Affiliation(s)
- Agnieszka Kubicka-Trząska
- Uniwersytet Jagielloński Collegium Medicum, Wydział Lekarski, Katedra Okulistyki, Klinika Okulistyki i Onkologii Okulistycznej Szpitala Uniwersyteckiego w Krakowie
| | - Izabella Karska-Basta
- Uniwersytet Jagielloński Collegium Medicum, Wydział Lekarski, Katedra Okulistyki, Klinika Okulistyki i Onkologii Okulistycznej Szpitala Uniwersyteckiego w Krakowie
| | - Katarzyna Żuber-Łaskawiec
- Uniwersytet Jagielloński Collegium Medicum, Wydział Lekarski, Katedra Okulistyki, Klinika Okulistyki i Onkologii Okulistycznej Szpitala Uniwersyteckiego w Krakowie
| |
Collapse
|
26
|
Cytoprotective Effects of Natural Highly Bio-Available Vegetable Derivatives on Human-Derived Retinal Cells. Nutrients 2020; 12:nu12030879. [PMID: 32214021 PMCID: PMC7146218 DOI: 10.3390/nu12030879] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 03/19/2020] [Accepted: 03/21/2020] [Indexed: 01/02/2023] Open
Abstract
Retinal pigment epithelial cells are crucial for retina maintenance, making their cytoprotection an excellent way to prevent or slow down retinal degeneration. In addition, oxidative stress, inflammation, apoptosis, neovascularization, and/or autophagy are key pathways involved in degenerative mechanisms. Therefore, here we studied the effects of curcumin, lutein, and/or resveratrol on human retinal pigment epithelial cells (ARPE-19). Cells were incubated with individual or combined agent(s) before induction of (a) H2O2-induced oxidative stress, (b) staurosporin-induced apoptosis, (c) CoCl2-induced hypoxia, or (d) a LED-autophagy perturbator. Metabolic activity, cellular survival, caspase 3/7 activity (casp3/7), cell morphology, VEGF levels, and autophagy process were assessed. H2O2 provoked a reduction in cell survival, whereas curcumin reduced metabolic activity which was not associated with cell death. Cell death induced by H2O2 was significantly reduced after pre-treatment with curcumin and lutein, but not resveratrol. Staurosporin increased caspase-3/7 activity (689%) and decreased cell survival by 32%. Curcumin or lutein protected cells from death induced by staurosporin. Curcumin, lutein, and resveratrol were ineffective on the increase of caspase 3/7 induced by staurosporin. Pre-treatment with curcumin or lutein prevented LED-induced blockage of autophagy flux. Basal-VEGF release was significantly reduced by lutein. Therefore, lutein and curcumin showed beneficial protective effects on human-derived retinal cells against several insults.
Collapse
|
27
|
Behnke V, Wolf A, Langmann T. The role of lymphocytes and phagocytes in age-related macular degeneration (AMD). Cell Mol Life Sci 2020; 77:781-788. [PMID: 31897541 PMCID: PMC11104950 DOI: 10.1007/s00018-019-03419-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 12/04/2019] [Accepted: 12/10/2019] [Indexed: 12/11/2022]
Abstract
Age-related macular degeneration (AMD) is a leading cause of visual impairment of the elderly population. Since AMD is a multifactorial age-related disease with various genetic risk factors, the understanding of its complex pathophysiology is still limited. However, animal experiments, genome-wide association data and the molecular profiling of AMD patient samples have highlighted a key role of systemic and local immune processes that contribute to this chronic eye disease. In this overview article, we concentrate on the role of lymphocytes and mononuclear phagocytes and their interplay in triggering a persistent immune response in the AMD retina. We preferentially review findings from human immune cell analyses and complement these with related findings in experimental models. We conclude that both immune cell types as their signaling network may be a rich source to identify novel molecular targets for immunomodulation in AMD.
Collapse
Affiliation(s)
- Verena Behnke
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931, Cologne, Germany
| | - Anne Wolf
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 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, 50931, Cologne, Germany.
- Center for Molecular Medicine Cologne (CMMC), 50931, Cologne, Germany.
| |
Collapse
|
28
|
Miao Q, Xu Y, Yin H, Zhang H, Ye J. KRT8 phosphorylation regulates the epithelial-mesenchymal transition in retinal pigment epithelial cells through autophagy modulation. J Cell Mol Med 2020; 24:3217-3228. [PMID: 32022439 PMCID: PMC7077598 DOI: 10.1111/jcmm.14998] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 12/15/2019] [Accepted: 12/24/2019] [Indexed: 01/13/2023] Open
Abstract
Proliferative vitreoretinopathy (PVR) is a severe ocular disease which results in complex retinal detachment and irreversible vision loss. The epithelial-mesenchymal transition (EMT) of retinal pigment epithelial (RPE) cells is considered to be critical in the pathogenesis of PVR. In this study, we focused on the potential impact of keratin 8 (KRT8) phosphorylation and autophagy on TGF-β2-induced EMT of RPE cells and explored the relationship between them. Using immunofluorescence and Western blot analysis, the co-localization of KRT8 and autophagy marker, as well as the abundance of phosphorylated KRT8 (p-KRT8) expression, was observed within subretinal and epiretinal membranes from PVR patients. Moreover, during TGF-β2-induced EMT process, we found that p-KRT8 was enhanced in RPE cells, which accompanied by an increase in autophagic flux. Inhibition of autophagy with pharmacological inhibitors or specific siRNAs was associated with a reduction in cell migration and the synthesis of several EMT markers. In the meantime, we demonstrated that p-KRT8 was correlated with the autophagy progression during the EMT of RPE cells. Knockdown the expression or mutagenesis of the critical phosphorylated site of KRT8 would induce autophagy impairment, through affecting the fusion of autophagosomes and lysosomes. Therefore, this study may provide a new insight into the pathogenesis of PVR and suggests the potential therapeutic value of p-KRT8 in the prevention and treatment of PVR.
Collapse
Affiliation(s)
- Qi Miao
- Eye Center, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yufeng Xu
- Eye Center, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Houfa Yin
- Eye Center, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Huina Zhang
- Eye Center, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Juan Ye
- Eye Center, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
29
|
Resveratrol as Inducer of Autophagy, Pro-Survival, and Anti-Inflammatory Stimuli in Cultured Human RPE Cells. Int J Mol Sci 2020; 21:ijms21030813. [PMID: 32012692 PMCID: PMC7036848 DOI: 10.3390/ijms21030813] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 12/31/2019] [Accepted: 01/07/2020] [Indexed: 11/17/2022] Open
Abstract
Purpose: To investigate the mechanism by which resveratrol acts upon retinal pigment epithelial (RPE) cells and to characterize its effect upon autophagy, survival, and inflammation, with consequent implications to treatment for age-related macular degeneration (AMD). Methods: Cultured ARPE-19 cells were exposed to 10 and 50 μM resveratrol. Cell survival/death was determined by annexin-FITC/propidium iodide using flow cytometry, while autophagy was studied by detecting autophagic vacuoles formation (acridine orange and transmission electron microscopy), as well as LC3II/I ratio and p62 expression by Western blot. In addition, time-lapse confocal microscopy of a pDENDRA-LC3 expression vector was performed to detect autophagy in transfected ARPE-19 cells under the different treatment conditions. Inhibition of proteasomal and autophagy-lysosomal fusion was carried out by MG-132 and chloroquine, respectively, while induction of autophagy was achieved by rapamycin treatment. Detection of secreted cytokines by ARPE-19 cells using Human XL Cytokine Array was performed under oxidative stress (H2O2) and resveratrol treatments, respectively. Results: Resveratrol induced autophagy in ARPE-19 cells as determined by augmented presence of autophagic vacuoles, increased LC3II/I ratio and decreased p62 expression, as well as time-lapse confocal microscopy using pDENDRA-LC3 expression vector. Resveratrol acted similarly to proteasomal inhibition and downstream of mammalian target of rapamycin (mTOR), since upstream inhibition of autophagy by 3-methyladenine could not inhibit autophagy in ARPE-19 cells. Co-treatmeant by rapamycin and/or proteasome inhibition showed no additive effect upon autophagy induction. ARPE-19 cells treated by resveratrol showed lower cell death rate compared to untreated controls. Resveratrol induced a specific anti-inflammatory response in ARPE-19 cells. Conclusions: Resveratrol can induce autophagy, pro-survival, and anti-inflammatory stimuli in ARPE-19 cells, properties which could be plausible to formulate future treatment modalities for AMD.
Collapse
|
30
|
Galimberti VE, Rothlin CV, Ghosh S. Funerals and Feasts: The Immunological Rites of Cell Death. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2019; 92:663-674. [PMID: 31866781 PMCID: PMC6913811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The immune system functions as a vanguard against pathogens and toxins. While it is mostly considered to be activated on the basis of self versus non-self recognition, injury/infection and damage are unavoidably associated with cell death. Does cell death play a role in the regulation of the immune response? Cell death, for better or for worse, is an omnipresent process in all stages of life that are observed throughout most tissues in multicellular organisms. From development to homeostasis in adult organisms, cells commit to scheduled death, while cases of injury and infection result in unscheduled cell death. Novel understanding of the molecular mechanisms that govern cell death demonstrate that, in fact, a plethora of molecular processes participate in directed dying. Parallel to the molecular modalities directing cell death are machineries employed by the organism to respond to dying cells, including either eliciting an inflammatory or immunological response or altogether avoiding it. Disturbing the careful coupling of these two processes is often met with pathology - on one hand a failure to respond to cell death may contribute to the lack of proper immune response or defective development, and on the other hand exaggerated or aberrant response to cell death can trigger unregulated inflammation, autoimmunity, or fibrosis/scarring. Here we review the molecular mechanisms and associated effector responses that accompany some of the most well-known cell death modalities - with an emphasis on efferocytosis, a process by which the dead cell is recognized and engulfed. In doing so, we highlight the TAM (TYRO3, AXL, MERTK) family of receptor tyrosine kinases (RTKs) that functions dually in the recognition and engulfment of dead cells, and as an important negative regulator of inflammation.
Collapse
Affiliation(s)
- Veronica E. Galimberti
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT,Department of Neurology, Yale University School of Medicine, New Haven, CT,To whom all correspondence should be addressed: Veronica E. Galimberti, Department of Immunobiology and Neurology, Yale University School of Medicine, New Haven, CT; Tel: 603-362-239,
| | - Carla V. Rothlin
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT,Department of Pharmacology, Yale University School of Medicine, New Haven, CT
| | - Sourav Ghosh
- Department of Neurology, Yale University School of Medicine, New Haven, CT,Department of Pharmacology, Yale University School of Medicine, New Haven, CT
| |
Collapse
|
31
|
Autophagy, lysosome dysfunction and mTOR inhibition in MNU-induced photoreceptor cell damage. Tissue Cell 2019; 61:98-108. [PMID: 31759414 DOI: 10.1016/j.tice.2019.09.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 09/21/2019] [Accepted: 09/23/2019] [Indexed: 01/03/2023]
Abstract
Progressive photoreceptor death is the main cause of retinal degeneration diseases. Determining the underlying mechanism of this process is essential for therapy improvement. Autophagy has long been considered to be involved in neuronal degeneration diseases, and the regulation of autophagy is thought to have potential implications for neurodegenerative disease therapies. However, whether autophagy is protective or destructive varies among diseases and is controversial. In the present study, we established an N-methyl-N-nitrosourea (MNU)-induced photoreceptor cell damage model in vitro that faithfully replicated photoreceptor cell death in retinal degeneration diseases. Cell viability was tested by 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxy-methoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium (MTS) assays. Reactive oxygen species (ROS) levels were assessed through 2,7-dichlorodihydrofluorescein diacetate (DCFH-DA) fluorescence. Autophagy was confirmed by observing autophagosomes using transmission electron microscopy (TEM). A lysosome tracker was used to identify acidic lysosomes in cells. We also measured the expression of some proteins related to autophagy, apoptosis and lysosomal degradation by western blot and immunofluorescence assays. We found that MNU could decrease photoreceptor cell viability in a time- and dose-dependent manner, and this change was accompanied by concomitant increases in ROS and the expression of the apoptosis-inducing protein cleaved caspase-3. Moreover, autophagy was activated by MNU treatment during this process. Inhibition of autophagy with 3-methyladenine accelerated cell damage. Lysosome dysfunction was confirmed by autophagosome enlargement and increased cathepsin expression, which was accompanied by mTOR dephosphorylation. In conclusion, autophagy was activated through inhibition of the PI3K/mTOR pathway in the context of MNU-induced photoreceptor cell death. Prolonged mTOR dephosphorylation and autophagy activation resulted in autophagic vacuole accumulation, as indicated by inefficient degradation in lysosomes, and further led to apoptosis.
Collapse
|
32
|
Li R, Du JH, Yao GM, Yao Y, Zhang J. Autophagy: a new mechanism for regulating VEGF and PEDF expression in retinal pigment epithelium cells. Int J Ophthalmol 2019; 12:557-562. [PMID: 31024806 DOI: 10.18240/ijo.2019.04.05] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 01/22/2019] [Indexed: 12/28/2022] Open
Abstract
AIM To investigate the regulation of vascular endothelial growth factors (VEGF) and pigment epithelium-derived factor (PEDF) expression by autophagy in retinal pigment epithelium (RPE) cells on exposure to hypoxia. METHODS ARPE-19, an RPE cell line, was treated as following: the control group was kept in a normoxic incubator; the hypoxia group was incubated in a hypoxic incubator with 1% O2/5% CO2/94% N2 for 24h; the hypoxia + 3-methyladenine (3-MA) group was pretreated with 10 mmol/L 3-MA for 1h and then in the hypoxic incubator for 24h; and the hypoxia + chloroquine (CQ) group was pretreated with 50 µmol/L CQ for 1h and then in the hypoxic incubator for 24h. The morphology and ultrastructure of the cells was observed by an inverted microscope or a transmission electronic microscope (TEM). Western blot was performed to assay the expression of autophagy-associated markers, including microtubule associated protein 1 light chain 3 B (LC3B), Beclin-1, Atg5 and p62. The concentration of VEGF and PEDF in the culture supernatant was determined by ELISA, and the ratio of VEGF/PEDF was calculated. RESULTS There were no obvious differences in cell morphology among different groups and autolysosomes could be observed in the cytoplasm in all groups. Compared to the control cells, the LC3B-II/I ratio and levels of Beclin-1 and Atg5 were significantly increased and p62 level was significantly decreased in the hypoxia group. With the increase of VEGF and decrease of PEDF concentration, the VEGF/PEDF ratio was significantly increased in the hypoxia group compared to the control cells. The LC3B-II/I ratio was significantly reduced by 3-MA treatment and increased by CQ treatment. The expressions of Beclin-1 and Atg5 were significantly reduced by 3-MA or CQ treatment, while expression of p62 was increased in the 3-MA or CQ treated cells. The concentration of VEGF was significantly decreased and PEDF increased, thereby the VEGF/PEDF ratio was decreased in the hypoxia + 3-MA group and hypoxia + CQ group compared with that in the hypoxia group. CONCLUSION Hypoxia leads to elevated autophagy in RPE cells, and expression of VEGF and PEDF might be regulated by autophagy on exposure to hypoxia to further participate in regulating the formation of retinal neovascularization.
Collapse
Affiliation(s)
- Rong Li
- Department of Ophthalmology, the First Affiliated Hospital of Xi'an Medical University, Xi'an 710077, Shaanxi Province, China
| | - Jun-Hui Du
- Department of Ophthalmology, Xi'an Ninth Hospital Affiliated to Medical College of Xi'an Jiaotong University, Xi'an 710054, Shaanxi Province, China
| | - Guo-Min Yao
- Department of Ophthalmology, the First Affiliated Hospital of Xi'an Medical University, Xi'an 710077, Shaanxi Province, China
| | - Yang Yao
- Department of Central Laboratory, the First Affiliated Hospital of Xi'an Medical University, Xi'an 710077, Shaanxi Province, China
| | - Jin Zhang
- Department of Ophthalmology, the First Hospital of Yulin, Yulin 719000, Shaanxi Province, China
| |
Collapse
|
33
|
Szatmári-Tóth M, Ilmarinen T, Mikhailova A, Skottman H, Kauppinen A, Kaarniranta K, Kristóf E, Lytvynchuk L, Veréb Z, Fésüs L, Petrovski G. Human Embryonic Stem Cell-Derived Retinal Pigment Epithelium-Role in Dead Cell Clearance and Inflammation. Int J Mol Sci 2019; 20:ijms20040926. [PMID: 30791639 PMCID: PMC6412543 DOI: 10.3390/ijms20040926] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 12/19/2018] [Accepted: 02/13/2019] [Indexed: 12/19/2022] Open
Abstract
Inefficient removal of dying retinal pigment epithelial (RPE) cells by professional phagocytes can result in debris formation and development of age-related macular degeneration (AMD). Chronic oxidative stress and inflammation play an important role in AMD pathogenesis. Only a few well-established in vitro phagocytosis assay models exist. We propose human embryonic stem cell-derived-RPE cells as a new model for studying RPE cell removal by professional phagocytes. The characteristics of human embryonic stem cells-derived RPE (hESC-RPE) are similar to native RPEs based on their gene and protein expression profile, integrity, and barrier properties or regarding drug transport. However, no data exist about RPE death modalities and how efficiently dying hESC-RPEs are taken upby macrophages, and whether this process triggers an inflammatory responses. This study demonstrates hESC-RPEs can be induced to undergo anoikis or autophagy-associated cell death due to extracellular matrix detachment or serum deprivation and hydrogen-peroxide co-treatment, respectively, similar to primary human RPEs. Dying hESC-RPEs are efficiently engulfed by macrophages which results in high amounts of IL-6 and IL-8 cytokine release. These findings suggest that the clearance of anoikic and autophagy-associated dying hESC-RPEs can be used as a new model for investigating AMD pathogenesis or for testing the in vivo potential of these cells in stem cell therapy.
Collapse
Affiliation(s)
- Mária Szatmári-Tóth
- Department of Biochemistry and Molecular Biology, University of Debrecen, Faculty of Medicine, 4032 Debrecen, Hungary.
| | - Tanja Ilmarinen
- Tampere University, Faculty of Medicine and Health Technology, 33014 Tampere, Finland.
| | - Alexandra Mikhailova
- Tampere University, Faculty of Medicine and Health Technology, 33014 Tampere, Finland.
| | - Heli Skottman
- Tampere University, Faculty of Medicine and Health Technology, 33014 Tampere, Finland.
| | - Anu Kauppinen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, 70211 Kuopio, Finland.
| | - Kai Kaarniranta
- Department of Ophthalmology, Institute of Clinical Medicine, University of Eastern Finland, 70211 Kuopio, Finland.
- Department of Ophthalmology, Kuopio University Hospital, 70029 Kuopio, Finland.
| | - Endre Kristóf
- Department of Biochemistry and Molecular Biology, University of Debrecen, Faculty of Medicine, 4032 Debrecen, Hungary.
| | - Lyubomyr Lytvynchuk
- Department of Ophthalmology, Justus-Liebig-University Giessen, Eye Clinic, University Hospital Giessen and Marburg GmbH, Campus Giessen, 35390 Giessen, Germany.
| | - Zoltán Veréb
- Department of Ophthalmology, Faculty of Medicine, University of Szeged, 6720 Szeged, Hungary.
| | - László Fésüs
- Department of Biochemistry and Molecular Biology, University of Debrecen, Faculty of Medicine, 4032 Debrecen, Hungary.
| | - Goran Petrovski
- Department of Biochemistry and Molecular Biology, University of Debrecen, Faculty of Medicine, 4032 Debrecen, Hungary.
- Department of Ophthalmology, Faculty of Medicine, University of Szeged, 6720 Szeged, Hungary.
- Center for Eye Research, Department of Ophthalmology, Oslo University Hospital and University of Oslo, Kirkeveien 166, 0450 Oslo, Norway.
| |
Collapse
|
34
|
Askou AL, Alsing S, Benckendorff JNE, Holmgaard A, Mikkelsen JG, Aagaard L, Bek T, Corydon TJ. Suppression of Choroidal Neovascularization by AAV-Based Dual-Acting Antiangiogenic Gene Therapy. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 16:38-50. [PMID: 30825671 PMCID: PMC6393707 DOI: 10.1016/j.omtn.2019.01.012] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 01/28/2019] [Accepted: 01/28/2019] [Indexed: 01/15/2023]
Abstract
Vascular endothelial growth factor A (VEGFA) is involved in the pathogenesis of vasoproliferative retinal diseases, such as exudative age-related macular degeneration (AMD). The objective of this study was to investigate whether dual-acting therapy based on the simultaneous expression of anti-VEGFA microRNAs (miRNAs) and the secreted, antiangiogenic protein pigment endothelial-derived factor (PEDF) delivered by adeno-associated virus (AAV) vectors provides improved protection against choroidal neovascularization (CNV). To investigate this, a multigenic AAV vector allowing retina pigment epithelium (RPE)-specific expression of anti-VEGFA miRNAs and PEDF was engineered. Robust expression of PEDF, driven by the RPE-specific vitelliform macular dystrophy 2 promoter, was observed in human cells and in mouse retina. A significant reduction in CNV was observed in a laser-induced CNV mouse model 57 days post-injection of the AAV5 particles conveying either anti-VEGFA miRNA and PEDF dual therapy or anti-VEGFA miRNA monotherapy. Overall, CNV reduction was most prominent in animals receiving dual-acting therapy. In both cases, the reduction in CNV was accompanied by a significant attenuation of VEGFA. In conclusion, the presented data reveal that gene therapy targeting VEGFA via multigenic AAV vectors displays combined efficacy, suggesting that dual-acting therapy is an important tool in future eye gene therapy for the treatment of neovascular ocular diseases, including AMD.
Collapse
Affiliation(s)
- Anne Louise Askou
- Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Sidsel Alsing
- Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | | | - Andreas Holmgaard
- Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | | | - Lars Aagaard
- Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Toke Bek
- Department of Ophthalmology, Aarhus University Hospital, 8000 Aarhus C, Denmark
| | - Thomas J Corydon
- Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark; Department of Ophthalmology, Aarhus University Hospital, 8000 Aarhus C, Denmark.
| |
Collapse
|
35
|
Chen X, Pan Z, Fang Z, Lin W, Wu S, Yang F, Li Y, Fu H, Gao H, Li S. Omega-3 polyunsaturated fatty acid attenuates traumatic brain injury-induced neuronal apoptosis by inducing autophagy through the upregulation of SIRT1-mediated deacetylation of Beclin-1. J Neuroinflammation 2018; 15:310. [PMID: 30409173 PMCID: PMC6225685 DOI: 10.1186/s12974-018-1345-8] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 10/24/2018] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Enhancing autophagy after traumatic brain injury (TBI) may decrease the expression of neuronal apoptosis-related molecules. Autophagy-mediated neuronal survival is regulated by the sirtuin family of proteins (SIRT). Omega-3 polyunsaturated fatty acids (ω-3 PUFA) are known to have antioxidative and anti-inflammatory effects. We previously demonstrated that ω-3 PUFA supplementation attenuated neuronal apoptosis by modulating the neuroinflammatory response through SIRT1-mediated deacetylation of the HMGB1/NF-κB pathway, leading to neuroprotective effects following experimental traumatic brain injury (TBI). However, no studies have elucidated if the neuroprotective effects of ω-3 PUFAs against TBI-induced neuronal apoptosis are modulated by SIRT1-mediated deacetylation of the autophagy pathway. METHODS The Feeney DM TBI model was adopted to induce TBI rats. Modified neurological severity scores, the rotarod test, brain water content, and Nissl staining were employed to determine the neuroprotective effects of ω-3 PUFA supplementation. Immunofluorescent staining and western blot analysis were used to detect Beclin-1 nuclear translocation and autophagy pathway activation. The impact of SIRT1 deacetylase activity on Beclin-1 acetylation and the interaction between cytoplasmic Beclin-1 and Bcl-2 were assessed to evaluate the neuroprotective effects of ω-3 PUFAs and to determine if these effects were dependent on SIRT1-mediated deacetylation of the autophagy pathway in order to gain further insight into the mechanisms underlying the development of neuroprotection after TBI. RESULTS ω-3 PUFA supplementation protected neurons against TBI-induced neuronal apoptosis via enhancement of the autophagy pathway. We also found that treatment with ω-3 PUFA significantly increased the NAD+/NADH ratio and SIRT1 activity following TBI. In addition, ω-3 PUFA supplementation increased Beclin-1 deacetylation and its nuclear export and induced direct interactions between cytoplasmic Beclin-1 and Bcl-2 by increasing SIRT1 activity following TBI. These events led to the inhibition of neuronal apoptosis and to neuroprotective effects through enhancing autophagy after TBI, possibly due to elevated SIRT1. CONCLUSIONS ω-3 PUFA supplementation attenuated TBI-induced neuronal apoptosis by inducing the autophagy pathway through the upregulation of SIRT1-mediated deacetylation of Beclin-1.
Collapse
Affiliation(s)
- Xiangrong Chen
- Department of Neurosurgery, The Second Affiliated Hospital, Fujian Medical University, Quanzhou, 362000, Fujian Province, China
| | - Zhigang Pan
- Department of Neurosurgery, The Second Affiliated Hospital, Fujian Medical University, Quanzhou, 362000, Fujian Province, China
| | - Zhongning Fang
- Department of Neurosurgery, The Second Affiliated Hospital, Fujian Medical University, Quanzhou, 362000, Fujian Province, China
| | - Weibin Lin
- Department of Neurosurgery, The Second Affiliated Hospital, Fujian Medical University, Quanzhou, 362000, Fujian Province, China
| | - Shukai Wu
- Department of Neurosurgery, The Second Affiliated Hospital, Fujian Medical University, Quanzhou, 362000, Fujian Province, China
| | - Fuxing Yang
- Department of Neurosurgery, The Second Affiliated Hospital, Fujian Medical University, Quanzhou, 362000, Fujian Province, China
| | - Yasong Li
- Department of Neurosurgery, The Second Affiliated Hospital, Fujian Medical University, Quanzhou, 362000, Fujian Province, China
| | - Huangde Fu
- Department of Neurosurgery, Affiliated Hospital of YouJiang Medical University for Nationalities, Baise, 533000, Guangxi Province, China.
| | - Hongzhi Gao
- Department of Neurosurgery, The Second Affiliated Hospital, Fujian Medical University, Quanzhou, 362000, Fujian Province, China.
| | - Shun Li
- Department of Neurosurgery, Affiliated Hospital of North Sichuan Medical College, Sichuan Province, Nanchong, 637000, China.
| |
Collapse
|
36
|
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
|
37
|
Qian M, Liu Z, Peng L, Tang X, Meng F, Ao Y, Zhou M, Wang M, Cao X, Qin B, Wang Z, Zhou Z, Wang G, Gao Z, Xu J, Liu B. Boosting ATM activity alleviates aging and extends lifespan in a mouse model of progeria. eLife 2018; 7:34836. [PMID: 29717979 PMCID: PMC5957528 DOI: 10.7554/elife.34836] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 04/16/2018] [Indexed: 02/06/2023] Open
Abstract
DNA damage accumulates with age (Lombard et al., 2005). However, whether and how robust DNA repair machinery promotes longevity is elusive. Here, we demonstrate that ATM-centered DNA damage response (DDR) progressively declines with senescence and age, while low dose of chloroquine (CQ) activates ATM, promotes DNA damage clearance, rescues age-related metabolic shift, and prolongs replicative lifespan. Molecularly, ATM phosphorylates SIRT6 deacetylase and thus prevents MDM2-mediated ubiquitination and proteasomal degradation. Extra copies of Sirt6 extend lifespan in Atm-/- mice, with restored metabolic homeostasis. Moreover, the treatment with CQ remarkably extends lifespan of Caenorhabditis elegans, but not the ATM-1 mutants. In a progeria mouse model with low DNA repair capacity, long-term administration of CQ ameliorates premature aging features and extends lifespan. Thus, our data highlights a pro-longevity role of ATM, for the first time establishing direct causal links between robust DNA repair machinery and longevity, and providing therapeutic strategy for progeria and age-related metabolic diseases. As cells live and divide, their genetic material gets damaged. The DNA damage response is a network of proteins that monitor, recognize and fix the damage, which is also called DNA lesions. For example, an enzyme called ATM senses when DNA is broken and then begins a process that will get it repaired, while another enzyme known as SIRT6 participates in the actual mending process. When organisms get older, the repair machinery becomes less efficient, and the number of DNA lesions and errors increases. The accumulation of DNA damage may cause the ‘symptoms’ of old age – from cancer, to wrinkles and the slowing down of the body’s chemical processes. In fact, individuals with defective ATMs (who thus struggle to repair their DNA) age abnormally fast; conversely, SIRT6 promotes longevity. If declining repair mechanisms cause aging, would boosting the DNA damage response slow down this process? Chloroquine is a drug used to combat malaria, but it can also enhance the activity of ATM without damaging DNA. Qian, Liu et al. show that chloroquine helps cells remove broken DNA and keep dividing for longer. In animals, the drug increases the lifespan of worms and prolongs the lives of mice who have mutations that make them age quicker. Qian, Liu et al. also demonstrate that ATM works by chemically altering the pro-longevity enzyme SIRT6. These changes make SIRT6 more stable, and keep it safe from cellular processes that destroy it. In addition, mice that are genetically engineered to lack ATM can survive for longer if they also produce extra SIRT6. These experiments show that enhancing the DNA damage response can slow down aging, thus linking the DNA repair machinery to longevity. Progeria is a group of rare genetic conditions with inefficient DNA repair; people with progeria age fast and die young. The results by Qian, Liu et al., if confirmed in humans, could provide a new way of treating these diseases.
Collapse
Affiliation(s)
- Minxian Qian
- Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Shenzhen University Health Science Center, Shenzhen, China.,Medical Research Center, Shenzhen University Health Science Center, Shenzhen, China.,Department of Biochemistry and Molecular Biology, Shenzhen University Health Science Center, Shenzhen, China
| | - Zuojun Liu
- Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Shenzhen University Health Science Center, Shenzhen, China.,Medical Research Center, Shenzhen University Health Science Center, Shenzhen, China.,Department of Biochemistry and Molecular Biology, Shenzhen University Health Science Center, Shenzhen, China
| | - Linyuan Peng
- Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Shenzhen University Health Science Center, Shenzhen, China.,Medical Research Center, Shenzhen University Health Science Center, Shenzhen, China.,Department of Biochemistry and Molecular Biology, Shenzhen University Health Science Center, Shenzhen, China
| | - Xiaolong Tang
- Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Shenzhen University Health Science Center, Shenzhen, China.,Medical Research Center, Shenzhen University Health Science Center, Shenzhen, China.,Department of Biochemistry and Molecular Biology, Shenzhen University Health Science Center, Shenzhen, China
| | - Fanbiao Meng
- Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Shenzhen University Health Science Center, Shenzhen, China.,Medical Research Center, Shenzhen University Health Science Center, Shenzhen, China.,Department of Biochemistry and Molecular Biology, Shenzhen University Health Science Center, Shenzhen, China
| | - Ying Ao
- Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Shenzhen University Health Science Center, Shenzhen, China.,Department of Biochemistry and Molecular Biology, Shenzhen University Health Science Center, Shenzhen, China
| | - Mingyan Zhou
- Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Shenzhen University Health Science Center, Shenzhen, China.,Medical Research Center, Shenzhen University Health Science Center, Shenzhen, China.,Department of Biochemistry and Molecular Biology, Shenzhen University Health Science Center, Shenzhen, China
| | - Ming Wang
- Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Shenzhen University Health Science Center, Shenzhen, China.,Medical Research Center, Shenzhen University Health Science Center, Shenzhen, China.,South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Xinyue Cao
- Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Shenzhen University Health Science Center, Shenzhen, China.,Medical Research Center, Shenzhen University Health Science Center, Shenzhen, China.,Department of Biochemistry and Molecular Biology, Shenzhen University Health Science Center, Shenzhen, China
| | - Baoming Qin
- South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Zimei Wang
- Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Shenzhen University Health Science Center, Shenzhen, China.,Department of Biochemistry and Molecular Biology, Shenzhen University Health Science Center, Shenzhen, China
| | - Zhongjun Zhou
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | - Guangming Wang
- East Hospital, Tongji University School of Medicine, Shanghai, China.,Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhengliang Gao
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Advanced Institute of Translational Medicine, Tongji University, Shanghai, China
| | - Jun Xu
- East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Baohua Liu
- Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Shenzhen University Health Science Center, Shenzhen, China.,Medical Research Center, Shenzhen University Health Science Center, Shenzhen, China.,Department of Biochemistry and Molecular Biology, Shenzhen University Health Science Center, Shenzhen, China
| |
Collapse
|
38
|
Piippo N, Korhonen E, Hytti M, Skottman H, Kinnunen K, Josifovska N, Petrovski G, Kaarniranta K, Kauppinen A. Hsp90 inhibition as a means to inhibit activation of the NLRP3 inflammasome. Sci Rep 2018; 8:6720. [PMID: 29712950 PMCID: PMC5928092 DOI: 10.1038/s41598-018-25123-2] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 04/12/2018] [Indexed: 02/07/2023] Open
Abstract
Once activated, the intracellular receptor NLRP3 assembles an inflammasome protein complex that facilitates the caspase-1-mediated maturation of IL-1β and IL-18. Inactive NLRP3 is guarded by a protein complex containing Hsp90. In response to stress stimuli, Hsp90 is released, and NLRP3 can be activated to promote inflammation. In this study, we blocked Hsp90 with geldanamycin and studied the fate of NLRP3 in human retinal pigment epithelial (RPE) cells. RPE cells play a central role in the development of age-related macular degeneration (AMD), a progressive eye disease causing severe vision loss in the elderly. IL-1α-primed ARPE-19 cells, human embryonal stem cell (hESC)-derived RPE cells, and primary human RPE cells were exposed to MG-132 and bafilomycin A to activate NLRP3 via the inhibition of proteasomes and autophagy, respectively. Additionally, RPE cells were treated with geldanamycin at different time points and the levels of NLRP3 and IL-1β were determined. Caspase-1 activity was measured using a commercial assay. Geldanamycin prevented the activation of the inflammasome in human RPE cells. NLRP3 released from its protective complex became degraded by autophagy or secreted from the cells. Controlled destruction of NLRP3 is a potential way to regulate the inflammation associated with chronic diseases, such as AMD.
Collapse
Affiliation(s)
- Niina Piippo
- School of Pharmacy, University of Eastern Finland, Kuopio, 70211, Finland
| | - Eveliina Korhonen
- School of Pharmacy, University of Eastern Finland, Kuopio, 70211, Finland
| | - Maria Hytti
- School of Pharmacy, University of Eastern Finland, Kuopio, 70211, Finland
| | - Heli Skottman
- Faculty of Medicine and Life Sciences, BioMediTech, University of Tampere, Tampere, 33014, Finland
| | - Kati Kinnunen
- Department of Ophthalmology, Kuopio University Hospital, Kuopio, 70211, Finland
| | - Natasha Josifovska
- Stem Cells and Eye Research Laboratory, Department of Ophthalmology, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, Szeged, Hungary
| | - Goran Petrovski
- Stem Cells and Eye Research Laboratory, Department of Ophthalmology, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, Szeged, Hungary.,Center for Eye Research, Department of Ophthalmology, Oslo University Hospital, University of Oslo, Oslo, Norway
| | - Kai Kaarniranta
- Department of Ophthalmology, Kuopio University Hospital, Kuopio, 70211, Finland.,Department of Ophthalmology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, 70211, Finland
| | - Anu Kauppinen
- School of Pharmacy, University of Eastern Finland, Kuopio, 70211, Finland.
| |
Collapse
|
39
|
Winiarczyk M, Kaarniranta K, Winiarczyk S, Adaszek Ł, Winiarczyk D, Mackiewicz J. Tear film proteome in age-related macular degeneration. Graefes Arch Clin Exp Ophthalmol 2018; 256:1127-1139. [PMID: 29696386 PMCID: PMC5956098 DOI: 10.1007/s00417-018-3984-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 03/18/2018] [Accepted: 04/09/2018] [Indexed: 01/05/2023] Open
Abstract
Purpose Age-related macular degeneration (AMD) is the main reason for blindness in elderly people in the developed countries. Current screening protocols have limitations in detecting the early signs of retinal degeneration. Therefore, it would be desirable to find novel biomarkers for early detection of AMD. Development of novel biomarkers would help in the prevention, diagnostics, and treatment of AMD. Proteomic analysis of tear film has shown promise in this research area. If an optimal set of biomarkers could be obtained from accessible body fluids, it would represent a reliable way to monitor disease progression and response to novel therapies. Methods Tear films were collected on Schirmer strips from a total of 22 patients (8 with wet AMD, 6 with dry AMD, and 8 control individuals). 2D electrophoresis was used to separate tear film proteins prior to their identification with matrix-assisted laser desorption/ionization time of flight spectrometer (MALDI-TOF/TOF) and matching with functional databases. Results A total of 342 proteins were identified. Most of them were previously described in various proteomic studies concerning AMD. Shootin-1, histatin-3, fidgetin-like protein 1, SRC kinase signaling inhibitor, Graves disease carrier protein, actin cytoplasmic 1, prolactin-inducible protein 1, and protein S100-A7A were upregulated in the tear film samples isolated from AMD patients and were not previously linked with this disease in any proteomic analysis. Conclusion The upregulated proteins supplement our current knowledge of AMD pathogenesis, providing evidence that certain specific proteins are expressed into the tear film in AMD. As far we are aware, this is the first study to have undertaken a comprehensive in-depth analysis of the human tear film proteome in AMD patients. Electronic supplementary material The online version of this article (10.1007/s00417-018-3984-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mateusz Winiarczyk
- Department of Vitreoretinal Surgery, Medical University of Lublin, Lublin, Poland
| | - Kai Kaarniranta
- Department of Ophthalmology, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Stanisław Winiarczyk
- Department of Epizootiology, University of Life Sciences of Lublin, Lublin, Poland
| | - Łukasz Adaszek
- Department of Epizootiology, University of Life Sciences of Lublin, Lublin, Poland
| | - Dagmara Winiarczyk
- Department of Epizootiology, University of Life Sciences of Lublin, Lublin, Poland
| | - Jerzy Mackiewicz
- Department of Vitreoretinal Surgery, Medical University of Lublin, Lublin, Poland.
| |
Collapse
|
40
|
Chen X, Wang H, Zhou M, Li X, Fang Z, Gao H, Li Y, Hu W. Valproic Acid Attenuates Traumatic Brain Injury-Induced Inflammation in Vivo: Involvement of Autophagy and the Nrf2/ARE Signaling Pathway. Front Mol Neurosci 2018; 11:117. [PMID: 29719500 PMCID: PMC5913341 DOI: 10.3389/fnmol.2018.00117] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 03/27/2018] [Indexed: 01/11/2023] Open
Abstract
Microglial activation and the inflammatory response in the central nervous system (CNS) play important roles in secondary damage after traumatic brain injury (TBI). Transcriptional activation of genes that limit secondary damage to the CNS are mediated by a cis-acting element called the antioxidant responsive element (ARE). ARE is known to associate with the transcription factor NF-E2-related factor 2 (Nrf2), a transcription factor that is associated with histone deacetylases (HDACs). This pathway, known as the Nrf2/ARE pathway, is a critical antioxidative factor pathway that regulates the balance of oxygen free radicals and the inflammatory response, and is also related to autophagic activities. Although valproic acid (VPA) is known to inhibit HDACs, it is unclear whether VPA plays a role in the microglia-mediated neuroinflammatory response after TBI via regulating oxidative stress and autophagy induced by the Nrf2/ARE signaling pathway. In this study, we demonstrate that microglial activation, oxidative stress, autophagy, and the Nrf2/ARE signaling pathway play essential roles in secondary injury following TBI. Treatment with VPA alleviated TBI-induced secondary brain injury, including neurological deficits, cerebral edema, and neuronal apoptosis. Moreover, VPA treatment upregulated the occurrence of autophagy and Nrf2/ARE pathway activity after TBI, and there was an increase in H3, H4 histone acetylation levels, accompanied by decreased transcriptional activity of the HDAC3 promoter in cortical lesions. These results suggest that VPA-mediated up-regulation of autophagy and antioxidative responses are likely due to increased activation of Nrf2/ARE pathway, through direct inhibition of HDAC3. This inhibition further reduces TBI-induced microglial activation and the subsequent inflammatory response, ultimately leading to neuroprotection.
Collapse
Affiliation(s)
- Xiangrong Chen
- Department of Neurosurgery, Jinling Hospital, Nanjing School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Department of Neurosurgery, The Second Affiliated Hospital, Fujian Medical University, Quanzhou, China
| | - Handong Wang
- Department of Neurosurgery, Jinling Hospital, Nanjing School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Mengliang Zhou
- Department of Neurosurgery, Jinling Hospital, Nanjing School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Xiang Li
- Department of Neurosurgery, Jinling Hospital, Nanjing School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Zhongning Fang
- Department of Neurosurgery, The Second Affiliated Hospital, Fujian Medical University, Quanzhou, China
| | - Hongzhi Gao
- Department of Neurosurgery, The Second Affiliated Hospital, Fujian Medical University, Quanzhou, China
| | - Yasong Li
- Department of Neurosurgery, The Second Affiliated Hospital, Fujian Medical University, Quanzhou, China
| | - Weipeng Hu
- Department of Neurosurgery, The Second Affiliated Hospital, Fujian Medical University, Quanzhou, China
| |
Collapse
|
41
|
Alhasani RH, Biswas L, Tohari AM, Zhou X, Reilly J, He JF, Shu X. Gypenosides protect retinal pigment epithelium cells from oxidative stress. Food Chem Toxicol 2018; 112:76-85. [DOI: 10.1016/j.fct.2017.12.037] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 12/06/2017] [Accepted: 12/19/2017] [Indexed: 02/07/2023]
|
42
|
Davies SP, Reynolds GM, Stamataki Z. Clearance of Apoptotic Cells by Tissue Epithelia: A Putative Role for Hepatocytes in Liver Efferocytosis. Front Immunol 2018; 9:44. [PMID: 29422896 PMCID: PMC5790054 DOI: 10.3389/fimmu.2018.00044] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 01/08/2018] [Indexed: 12/11/2022] Open
Abstract
Toxic substances and microbial or food-derived antigens continuously challenge the liver, which is tasked with their safe neutralization. This vital organ is also important for the removal of apoptotic immune cells during inflammation and has been previously described as a “graveyard” for dying lymphocytes. The clearance of apoptotic and necrotic cells is known as efferocytosis and is a critical liver function to maintain tissue homeostasis. Much of the research into this form of immunological control has focused on Kupffer cells, the liver-resident macrophages. However, hepatocytes (and other liver resident cells) are competent efferocytes and comprise 80% of the liver mass. Little is known regarding the mechanisms of apoptotic and necrotic cell capture by epithelia, which lack key receptors that mediate phagocytosis in macrophages. Herein, we discuss recent developments that increased our understanding of efferocytosis in tissues, with a special focus on the liver parenchyma. We discuss the impact of efferocytosis in health and in inflammation, highlighting the role of phagocytic epithelia.
Collapse
Affiliation(s)
- Scott P Davies
- Centre for Liver Research, College of Medical and Dental Sciences, Institute for Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Gary M Reynolds
- Centre for Liver Research, College of Medical and Dental Sciences, Institute for Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom.,Centre for Liver Research and National Institute for Health Research (NIHR) Birmingham Liver Biomedical Research Unit, College of Medical and Dental Sciences, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom.,University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Zania Stamataki
- Centre for Liver Research, College of Medical and Dental Sciences, Institute for Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
43
|
Microtubule-Associated Protein 1 Light Chain 3 (LC3) Isoforms in RPE and Retina. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1074:609-616. [PMID: 29721994 DOI: 10.1007/978-3-319-75402-4_74] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
Microtubule-associated protein 1 light chain 3 (MAP1LC3), a human homologue of yeast Atg8, is an essential component of autophagy. LC3 plays a critical role in hybrid degradation pathways in which some but not all components of autophagy are coupled with phagocytosis in a process known as LC3-associated phagocytosis (LAP). LC3 exists as three highly homologous isoforms in human (LC3A, LC3B, and LC3C) with two of these (LC3A and LC3B) in mouse. LC3B predominated in both fetal and adult human retinal pigment epithelium (RPE) relative to LC3A and LC3C, while in mouse RPE and neural retina, LC3A and LC3B were expressed at approximately equivalent levels. In situ hybridization studies localized LC3A and LC3B transcripts in the retina and RPE. LC3B protein was detected in C57Bl6/J RPE and retinal lysates and was absent in the LC3BKO mouse.
Collapse
|
44
|
Resveratrol reverses the adverse effects of bevacizumab on cultured ARPE-19 cells. Sci Rep 2017; 7:12242. [PMID: 28947815 PMCID: PMC5612947 DOI: 10.1038/s41598-017-12496-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 09/11/2017] [Indexed: 01/03/2023] Open
Abstract
Age-related macular degeneration (AMD) and proliferative diabetic retinopathy (PDR) are one of the major causes of blindness caused by neo-vascular changes in the retina. Intravitreal anti-VEGF injections are widely used in the treatment of wet-AMD and PDR. A significant percentage of treated patients have complications of repeated injections. Resveratrol (RES) is a polyphenol phytoalexin with anti-oxidative, anti-inflammatory and anti-proliferative properties. Hence, we hypothesized that if RES is used in combination with bevacizumab (BEV, anti-VEGF), it could reverse the adverse effects that precipitate fibrotic changes, drusen formation, tractional retinal detachment and so on. Human retinal pigment epithelial cells were treated with various combinations of BEV and RES. There was partial reduction in secreted VEGF levels compared to untreated controls. Epithelial-mesenchymal transition was lower in BEV + RES treated cultures compared to BEV treated cultures. The proliferation status was similar in BEV + RES as well as BEV treated cultures both groups. Phagocytosis was enhanced in the presence of BEV + RES compared to BEV. Furthermore, we observed that notch signaling was involved in reversing the adverse effects of BEV. This study paves way for a combinatorial strategy to treat as well as prevent adverse effects of therapy in patients with wet AMD and PDR.
Collapse
|
45
|
Hu L, Yang H, Ai M, Jiang S. Inhibition of TLR4 alleviates the inflammation and apoptosis of retinal ganglion cells in high glucose. Graefes Arch Clin Exp Ophthalmol 2017; 255:2199-2210. [DOI: 10.1007/s00417-017-3772-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 07/03/2017] [Accepted: 08/01/2017] [Indexed: 01/20/2023] Open
|
46
|
Arjamaa O, Aaltonen V, Piippo N, Csont T, Petrovski G, Kaarniranta K, Kauppinen A. Hypoxia and inflammation in the release of VEGF and interleukins from human retinal pigment epithelial cells. Graefes Arch Clin Exp Ophthalmol 2017. [PMID: 28631245 DOI: 10.1007/s00417-017-3711-0] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
PURPOSE Retinal diseases are closely associated with both decreased oxygenation and increased inflammation. It is not known if hypoxia-induced vascular endothelial growth factor (VEGF) expression in the retina itself evokes inflammation, or whether inflammation is a prerequisite for the development of neovascularization. METHODS Human ARPE-19 cell line and primary human retinal pigment epithelium (RPE) cells were used. ARPE-19 cells were kept either under normoxic (24 h or 48 h) or hypoxic conditions (1% O2, 24 h). Part of the cells were re-oxygenated (24 h). Some ARPE-19 cells were additionally pre-treated with bacterial lipopolysaccharide (LPS). The levels of IL-6, IL-8, IL-1β, and IL-18 were determined from medium samples by an enzyme-linked immunosorbent assay (ELISA) method. Primary human RPE cells were exposed to hypoxia for 24 h, and the subsequent release of IL-6 and IL-8 was measured with ELISA. VEGF secretion from ARPE-19 cells was determined up to 24 h. RESULTS Hypoxia induced significant (P < 0.01) increases in the levels of both IL-6 and IL-8 in ARPE-19 cells, and LPS pre-treatment further enhanced these responses. Hypoxia exposure did not affect the IL-1β or IL-18 release irrespective of LPS pre-treatment. If primary RPE cells were incubated for 4 h in hypoxic conditions, IL-6 and IL-8 concentrations were increased by 7 and 8-fold respectively. Hypoxia increased the VEGF secretion from ARPE-19 cells in a similar manner with or without pre-treatment with LPS. CONCLUSIONS Hypoxia causes an inflammatory reaction in RPE cells that is potentiated by pre-treatment with the Toll-like receptor-activating agent, LPS. The secretion of VEGF from these cells is regulated directly by hypoxia and is not mediated by inflammation.
Collapse
Affiliation(s)
- Olli Arjamaa
- Department of Ophthalmology, University of Turku, Turku, 20521, Finland.
| | - Vesa Aaltonen
- Department of Ophthalmology, University of Turku, Turku, 20521, Finland.,Department of Ophthalmology, Turku University Hospital, Turku, Finland
| | - Niina Piippo
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland.,Department of Ophthalmology, University of Eastern Finland, Kuopio, Finland
| | - Tamás Csont
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Goran Petrovski
- Center for Eye Research, Department of Ophthalmology, Oslo University Hospital, University of Oslo, Oslo, Norway.,Department of Ophthalmology, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, Szeged, Hungary
| | - Kai Kaarniranta
- Department of Ophthalmology, University of Eastern Finland, Kuopio, Finland.,Department of Ophthalmology, Kuopio University Hospital, Kuopio, Finland
| | - Anu Kauppinen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
47
|
Kaarniranta K, Petrovski G, Kauppinen A. The Nobel Prized cellular target autophagy in eye diseases. Acta Ophthalmol 2017; 95:335-336. [PMID: 28603953 DOI: 10.1111/aos.13344] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Kai Kaarniranta
- Department of Ophthalmology; University of Eastern Finland and Kuopio University Hospital; Kuopio Finland
| | - Goran Petrovski
- Stem Cells and Eye Research Laboratory; Department of Ophthalmology; Faculty of Medicine; University of Szeged; Szeged Hungary
- Department of Ophthalmology; Center for Eye Research; Oslo University Hospital; University of Oslo; Oslo Norway
| | - Anu Kauppinen
- Faculty of Health Sciences; School of Pharmacy; University of Eastern Finland; Kuopio Finland
| |
Collapse
|
48
|
Singh M, Tyagi SC. Homocysteine mediates transcriptional changes of the inflammatory pathway signature genes in human retinal pigment epithelial cells. Int J Ophthalmol 2017; 10:696-704. [PMID: 28546923 DOI: 10.18240/ijo.2017.05.06] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2016] [Accepted: 03/01/2017] [Indexed: 12/28/2022] Open
Abstract
AIM To test whether homocysteine (Hcy) can influence the transcriptional profile, we hypothesized that Hcy can lead to the induction of proinflammatory molecules in the retinal cells of aging people. METHODS An unbiased in vitro inflammatory pathway focused study was designed employing retinal pigment epithelial (RPE) cell line, ARPE-19. Cells were cultured in the presence or absence of Hcy to capture target genes' expression profile. Three different concentrations of Hcy were added in the culture medium of confluent monolayers. cRNAs were made from the isolated total RNAs and the labeled cRNA probes were hybridized to microarrays specific for human disease pathway inflammatory cytokines, chemokines and their receptor gene micro-array panels as per manufacture's recommendations. Two Hcy up-regulated molecules: IL6 and CEBPB were further validated via Western blot analysis. Hcy's effect on ARPE-19 cellular morphology and genomic DNA integrity were also evaluated. RESULTS Gene microarray analyses of RPE cells in response to Hcy treatment revealed alterations in the expressions of several inflammatory gene transcripts such as CCL5, CEBPB, IL13RA2, IL15RA, IL6, IL8 and CXCL3 that were up-regulated. The transcripts for C3, CCL2, IL11RA and IL18 genes exhibited down-regulation. The IL6 and CEBPB expressions were subsequently validated at the protein levels. Treatment of the retinal cells with increasing Hcy concentration influenced their density in culture however their morphology and DNA integrity remained unaffected. CONCLUSION These findings suggest that Hcy can potentially mediate the expression of chemokines, cytokines and interleukins receptors in the retinal cells without having any debilitating effects on their morphology and the genomic DNA integrity.
Collapse
Affiliation(s)
- Mahavir Singh
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Suresh C Tyagi
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| |
Collapse
|