1
|
Chang YC, Kao TE, Chen CL, Lin YC, Hwang DK, Hwang YS, Lin CJ, Chan WC, Lin CP, Chen SN, Sheu SJ. Use of corticosteroids in non-infectious uveitis - expert consensus in Taiwan. Ann Med 2024; 56:2352019. [PMID: 38747459 PMCID: PMC11097703 DOI: 10.1080/07853890.2024.2352019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 04/21/2024] [Indexed: 05/18/2024] Open
Abstract
PURPOSE To offer consensus on the utilization of corticosteroids (CS) for treating non-infectious uveitis in the context of clinical practice in Taiwan. This entails examining the different administration methods, their advantages and disadvantages, and considering alternative treatments according to the prevailing evidence and health policies. METHODS Ten ophthalmologists and one rheumatologist convened on December 11, 2022, to review and discuss literature on the topic. The databases explored were the Central Cochrane library, EMBASE, Medline, PUBMED, and Web of Science using relevant keywords. The search spanned from January 1996 to June 2023. After the initial results of the literature review were presented, open voting determined the final statements, with a statement being accepted if it secured more than 70% agreement. This consensus was then presented at significant meetings for further discussions before the final version was established. RESULTS A flow chart and nine statements emerged from the deliberations. They address the importance of CS in uveitis management, guidelines for using topical CS, indications for both periocular or intravitreal and systemic therapies, and tapering and discontinuation methods for both topical and systemic CS. CONCLUSION While CS are a cornerstone for non-infectious uveitis treatment, their administration requires careful consideration, depending on the clinical situation and the specific type of uveitis. The consensus generated from this article provides a guideline for practitioners in Taiwan, taking into account local health policies and the latest research on the subject. It emphasizes the significance of strategic tapering, the potential for alternative therapies, and the importance of patient-centric care.
Collapse
Affiliation(s)
- Yo-Chen Chang
- Department of Ophthalmology, School of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Ophthalmology, Kaohsiung Municipal Siaogang Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Ophthalmology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Tzu-En Kao
- Cheng Ching International Eye Hospital, Kaohsiung, Taiwan
| | - Ching-Long Chen
- Department of Ophthalmology, National Defense Medical Center, Taipei, Taiwan
| | - Yu-Chih Lin
- Division of Allergy, Immunology and Rheumatology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - De-Kuang Hwang
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yih-Shiou Hwang
- College of Medicine, Chang Gung University, Taoyuan City, Taiwan
- Department of Ophthalmology, Chang Gung Memorial Hospital, Taoyuan City, Taiwan
- Department of Ophthalmology, Chang Gung Memorial Hospital, Xiamen Branch, Xiamen, China
- Department of Ophthalmology, Jen-Ai Hospital Dali Branch, Taichung, Taiwan
- Department of Optometry, Asia University, Taichung, Taiwan
| | - Chun-Ju Lin
- Department of Optometry, Asia University, Taichung, Taiwan
- Department of Ophthalmology, China Medical University Hospital, China Medical University, Taichung, Taiwan
- School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan
| | - Wei-Chun Chan
- Department of Ophthalmology, Mackay Memorial Hospital, Taipei, Taiwan
| | - Chang-Ping Lin
- Department of Ophthalmology, National Taiwan University Hospital, Taipei, Taiwan
| | - San-Ni Chen
- Department of Ophthalmology, China Medical University Hospital, China Medical University, Taichung, Taiwan
- School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan
| | - Shwu-Jiuan Sheu
- Department of Ophthalmology, School of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Ophthalmology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| |
Collapse
|
2
|
Calpe E, Fernández-Engroba J, Julio G, Pavan J, Bonel C, Barraquer RI. Risk factors for early and late retinal detachment after boston type I keratoprosthesis surgery. Graefes Arch Clin Exp Ophthalmol 2024:10.1007/s00417-024-06613-8. [PMID: 39259299 DOI: 10.1007/s00417-024-06613-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/30/2024] [Accepted: 08/09/2024] [Indexed: 09/13/2024] Open
Abstract
PURPOSE To determine the differential risk factors for retinal detachment (RD) after Boston Type 1 Keratoprosthesis (B1KPro) during the first year after surgery (early RD; ERD) and afterwards (late RD; LRD). METHODS Retrospective cohort study of 94 eyes (90 patients) undergoing B1KPro implantation at Centro de Oftalmología Barraquer from June 2006 to July 2022 with a minimum follow-up of one year. RESULTS The incidence of RD in the whole sample after B1KPro implantation was 29% (27/94), occurring in 21% of eyes (ERD; 20/94) the first year, and in 7% afterwards (LRD; 7/94). Hypotony was revealed as a significant risk factor for ERD (16/19 = 84%; OR = 14.5, P < 0.0001), together with aphakia (16/20 = 80%; OR = 5.9, P = 0.004), intraocular lens (IOL) removal (4/8 = 50%; OR = 6.2, P = 0.03), previous choroidal detachment (CD; 7/20 = 35%; OR = 35.5, P = 0.001) and previous pars plana vitrectomy (PPV; 15/20 = 75%; OR = 6.6, P = 0.006). Multivariate analysis included hypotony and PPV at any time as variables significantly associated with ERD occurrence. The occurrence of LRD was significantly related to peripheral retinal changes, detected few days before (4/6 = 66%; OR = 65, P < 0.0001). CONCLUSION Hypotony emerged as one of the main factors influencing ERD occurrence after B1KPro implantation. Additionally, previous CD, PPV, aphakia, and IOL removal should be considered to estimate the risk of postoperative retinal detachment. In contrast, peripheral retinal changes after B1KPro seem to be significantly related to LRD occurrence. This study reinforces the need for frequent intraocular pressure evaluation, and comprehensive periodic retinal assessment over time. KEY MESSAGES What is known Retinal detachment (RD) is a severe complication after Boston Type 1 Keratoprosthesis (B1KPro) implantation, resulting in a permanent reduction of visual acuity in a clinically relevant percentage of eyes. The time of RD occurrence after B1KPro implantation differs in the literature but is reported to be more common within the first year. Previous aphakia, choroidal detachment, or RD repair, intraocular lens removal, vitritis, or history of Nd-YAG laser have been described as risk factors for RD after B1KPro surgery in samples with varying follow-ups. WHAT IS NEW Multivariate analysis in the first year after B1KPro surgery showed hypotony and pars plana vitrectomy at any time as significant risk factors for early RD. Late RD cases, occurring after one year following the B1KPro surgery, seem to be significantly related to peripheral retinal changes suffered a few days before loss of visual acuity and RD diagnosis. It is plausible to think that risk factors for RD after B1KPro differ in short and long postoperative times.
Collapse
Affiliation(s)
- Eva Calpe
- Centro de Oftalmología Barraquer, Barcelona, Spain
- Institut Universitari Barraquer, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Jorge Fernández-Engroba
- Centro de Oftalmología Barraquer, Barcelona, Spain
- Institut Universitari Barraquer, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Gemma Julio
- Centro de Oftalmología Barraquer, Barcelona, Spain.
- Institut Universitari Barraquer, Universitat Autònoma de Barcelona, Barcelona, Spain.
| | - Josip Pavan
- Department of Ophthalmology, Dubrava University Hospital, Zagreb, Croatia
| | - Clara Bonel
- Centro de Oftalmología Barraquer, Barcelona, Spain
- Institut Universitari Barraquer, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Rafael I Barraquer
- Centro de Oftalmología Barraquer, Barcelona, Spain
- Institut Universitari Barraquer, Universitat Autònoma de Barcelona, Barcelona, Spain
- Universitat Internacional de Catalunya, Barcelona, Spain
| |
Collapse
|
3
|
Lee SJ, Noh SE, Jo DH, Cho CS, Park KS, Kim JH. IL-10-induced modulation of macrophage polarization suppresses outer-blood-retinal barrier disruption in the streptozotocin-induced early diabetic retinopathy mouse model. FASEB J 2024; 38:e23638. [PMID: 38713098 DOI: 10.1096/fj.202400053r] [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: 01/10/2024] [Revised: 03/26/2024] [Accepted: 04/18/2024] [Indexed: 05/08/2024]
Abstract
Diabetic retinopathy (DR) is associated with ocular inflammation leading to retinal barrier breakdown, vascular leakage, macular edema, and vision loss. DR is not only a microvascular disease but also involves retinal neurodegeneration, demonstrating that pathological changes associated with neuroinflammation precede microvascular injury in early DR. Macrophage activation plays a central role in neuroinflammation. During DR, the inflammatory response depends on the polarization of retinal macrophages, triggering pro-inflammatory (M1) or anti-inflammatory (M2) activity. This study aimed to determine the role of macrophages in vascular leakage through the tight junction complexes of retinal pigment epithelium, which is the outer blood-retinal barrier (BRB). Furthermore, we aimed to assess whether interleukin-10 (IL-10), a representative M2-inducer, can decrease inflammatory macrophages and alleviate outer-BRB disruption. We found that modulation of macrophage polarization affects the structural and functional integrity of ARPE-19 cells in a co-culture system under high-glucose conditions. Furthermore, we demonstrated that intravitreal IL-10 injection induces an increase in the ratio of anti-inflammatory macrophages and effectively suppresses outer-BRB disruption and vascular leakage in a mouse model of early-stage streptozotocin-induced diabetes. Our results suggest that modulation of macrophage polarization by IL-10 administration during early-stage DR has a promising protective effect against outer-BRB disruption and vascular leakage. This finding provides valuable insights for early intervention in DR.
Collapse
Affiliation(s)
- Seok Jae Lee
- Fight against Angiogenesis-Related Blindness (FARB) Laboratory, Clinical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
- Global Excellence Center for Gene & Cell Therapy (GEC-GCT), Seoul National University Hospital, Seoul, Republic of Korea
| | - Sung-Eun Noh
- Fight against Angiogenesis-Related Blindness (FARB) Laboratory, Clinical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
- Global Excellence Center for Gene & Cell Therapy (GEC-GCT), Seoul National University Hospital, Seoul, Republic of Korea
| | - Dong Hyun Jo
- Global Excellence Center for Gene & Cell Therapy (GEC-GCT), Seoul National University Hospital, Seoul, Republic of Korea
- Department of Anatomy & Cell Biology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Chang Sik Cho
- Fight against Angiogenesis-Related Blindness (FARB) Laboratory, Clinical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
- Global Excellence Center for Gene & Cell Therapy (GEC-GCT), Seoul National University Hospital, Seoul, Republic of Korea
| | - Kyu-Sang Park
- Department of Physiology, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - Jeong Hun Kim
- Fight against Angiogenesis-Related Blindness (FARB) Laboratory, Clinical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
- Global Excellence Center for Gene & Cell Therapy (GEC-GCT), Seoul National University Hospital, Seoul, Republic of Korea
- Department of Biomedical Sciences & Ophthalmology, Seoul National University College of Medicine, Seoul, Republic of Korea
- Institute of Reproductive Medicine and Population, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
4
|
Kim H, Roh H, Kim SH, Lee K, Im M, Oh SJ. Effective protection of photoreceptors using an inflammation-responsive hydrogel to attenuate outer retinal degeneration. NPJ Regen Med 2023; 8:68. [PMID: 38097595 PMCID: PMC10721838 DOI: 10.1038/s41536-023-00342-y] [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: 04/22/2023] [Accepted: 11/30/2023] [Indexed: 12/17/2023] Open
Abstract
Retinitis pigmentosa (RP) is an outer retinal degenerative disease that can lead to photoreceptor cell death and profound vision loss. Although effective regulation of intraretinal inflammation can slow down the progression of the disease, an efficient anti-inflammatory treatment strategy is still lacking. This study reports the fabrication of a hyaluronic acid-based inflammation-responsive hydrogel (IRH) and its epigenetic regulation effects on retinal degeneration. The injectable IRH was designed to respond to cathepsin overexpression in an inflammatory environment. The epigenetic drug, the enhancer of zeste homolog 2 (EZH2) inhibitors, was loaded into the hydrogel to attenuate inflammatory factors. On-demand anti-inflammatory effects of microglia cells via the drug-loaded IRH were verified in vitro and in vivo retinal degeneration 10 (rd10) mice model. Therefore, our IRH not only reduced intraretinal inflammation but also protected photoreceptors morphologically and functionally. Our results suggest the IRH reported here can be used to considerably delay vision loss caused by RP.
Collapse
Affiliation(s)
- Hyerim Kim
- Program in Nanoscience and Technology, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826, South Korea
| | - Hyeonhee Roh
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, South Korea
- School of Electrical Engineering, College of Engineering, Korea University, Seoul, 02841, South Korea
| | - Sang-Heon Kim
- Center for Biomaterials, Biomedical Research Institute, KIST, Seoul, 02792, South Korea
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul, 02792, South Korea
| | - Kangwon Lee
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826, South Korea.
- Research Institute for Convergence Science, Seoul National University, Seoul, 08826, South Korea.
| | - Maesoon Im
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, South Korea.
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul, 02792, South Korea.
- KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul, 02447, South Korea.
| | - Seung Ja Oh
- Department of Genetics and Biotechnology, College of Life Sciences, Kyung Hee University, Yongin, 17104, South Korea.
| |
Collapse
|
5
|
Wei J, Mu J, Tang Y, Qin D, Duan J, Wu A. Next-generation nanomaterials: advancing ocular anti-inflammatory drug therapy. J Nanobiotechnology 2023; 21:282. [PMID: 37598148 PMCID: PMC10440041 DOI: 10.1186/s12951-023-01974-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 06/29/2023] [Indexed: 08/21/2023] Open
Abstract
Ophthalmic inflammatory diseases, including conjunctivitis, keratitis, uveitis, scleritis, and related conditions, pose considerable challenges to effective management and treatment. This review article investigates the potential of advanced nanomaterials in revolutionizing ocular anti-inflammatory drug interventions. By conducting an exhaustive analysis of recent advancements and assessing the potential benefits and limitations, this review aims to identify promising avenues for future research and clinical applications. The review commences with a detailed exploration of various nanomaterial categories, such as liposomes, dendrimers, nanoparticles (NPs), and hydrogels, emphasizing their unique properties and capabilities for accurate drug delivery. Subsequently, we explore the etiology and pathophysiology of ophthalmic inflammatory disorders, highlighting the urgent necessity for innovative therapeutic strategies and examining recent preclinical and clinical investigations employing nanomaterial-based drug delivery systems. We discuss the advantages of these cutting-edge systems, such as biocompatibility, bioavailability, controlled release, and targeted delivery, alongside potential challenges, which encompass immunogenicity, toxicity, and regulatory hurdles. Furthermore, we emphasize the significance of interdisciplinary collaborations among material scientists, pharmacologists, and clinicians in expediting the translation of these breakthroughs from laboratory environments to clinical practice. In summary, this review accentuates the remarkable potential of advanced nanomaterials in redefining ocular anti-inflammatory drug therapy. We fervently support continued research and development in this rapidly evolving field to overcome existing barriers and improve patient outcomes for ophthalmic inflammatory disorders.
Collapse
Affiliation(s)
- Jing Wei
- School of Ophthalmology, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Jinyu Mu
- School of Ophthalmology, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Yong Tang
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Education Ministry Key Laboratory of Medical Electrophysiology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Dalian Qin
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Education Ministry Key Laboratory of Medical Electrophysiology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Junguo Duan
- School of Ophthalmology, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China.
| | - Anguo Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Education Ministry Key Laboratory of Medical Electrophysiology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
6
|
Paskeviciute E, Chen M, Xu H, Honoré B, Vorum H, Sørensen TL, Christensen JP, Thomsen AR, Nissen MH, Steffensen MA. Systemic virus infection results in CD8 T cell recruitment to the retina in the absence of local virus infection. Front Immunol 2023; 14:1221511. [PMID: 37662932 PMCID: PMC10471971 DOI: 10.3389/fimmu.2023.1221511] [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: 05/12/2023] [Accepted: 07/31/2023] [Indexed: 09/05/2023] Open
Abstract
During recent years, evidence has emerged that immune privileged sites such as the CNS and the retina may be more integrated in the systemic response to infection than was previously believed. In line with this, it was recently shown that a systemic acute virus infection leads to infiltration of CD8 T cells in the brains of immunocompetent mice. In this study, we extend these findings to the neurological tissue of the eye, namely the retina. We show that an acute systemic virus infection in mice leads to a transient CD8 T cell infiltration in the retina that is not directed by virus infection inside the retina. CD8 T cells were found throughout the retinal tissue, and had a high expression of CXCR6 and CXCR3, as also reported for tissue residing CD8 T cells in the lung and liver. We also show that the pigment epithelium lining the retina expresses CXCL16 (the ligand for CXCR6) similar to epithelial cells of the lung. Thus, our results suggest that the retina undergoes immune surveillance during a systemic infection, and that this surveillance appears to be directed by mechanisms similar to those described for non-privileged tissues.
Collapse
Affiliation(s)
- Egle Paskeviciute
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Mei Chen
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queens University of Belfast, Belfast, Ireland
| | - Heping Xu
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queens University of Belfast, Belfast, Ireland
| | - Bent Honoré
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Henrik Vorum
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
- Department of Ophthalmology, Aalborg University Hospital, Aalborg, Denmark
| | - Torben Lykke Sørensen
- Department of Ophthalmology, Zealand University Hospital, Roskilde, Denmark
- Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Allan Randrup Thomsen
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Mogens Holst Nissen
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | | |
Collapse
|
7
|
Hu B, Ma JX, Duerfeldt AS. The cGAS-STING pathway in diabetic retinopathy and age-related macular degeneration. Future Med Chem 2023; 15:717-729. [PMID: 37166075 PMCID: PMC10194038 DOI: 10.4155/fmc-2022-0301] [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: 12/08/2022] [Accepted: 03/28/2023] [Indexed: 05/12/2023] Open
Abstract
Diabetic retinopathy and age-related macular degeneration are common retinal diseases with shared pathophysiology, including oxidative stress-induced inflammation. Cellular mechanisms responsible for converting oxidative stress into retinal damage are ill-defined but have begun to clarify. One common outcome of retinal oxidative stress is mitochondrial damage and subsequent release of mitochondrial DNA into the cytosol. This leads to activation of the cGAS-STING pathway, resulting in interferon release and disease-amplifying inflammation. This review summarizes the evolving link between aberrant cGAS-STING signaling and inflammation in common retinal diseases and provides prospective for targeting this system in diabetic retinopathy and age-related macular degeneration. Further defining the roles of this system in the retina is expected to reveal new disease pathology and novel therapeutic approaches.
Collapse
Affiliation(s)
- Bo Hu
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, Minneapolis, MN 55414, USA
| | - Jian-Xing Ma
- Department of Biochemistry, Wake Forest University School of Medicine, Winston Salem, NC 27101, USA
| | - Adam S Duerfeldt
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, Minneapolis, MN 55414, USA
| |
Collapse
|
8
|
Jeong S, Shin EC, Lee JH, Ha JH. Particulate Matter Elevates Ocular Inflammation and Endoplasmic Reticulum Stress in Human Retinal Pigmented Epithelium Cells. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:4766. [PMID: 36981676 PMCID: PMC10049273 DOI: 10.3390/ijerph20064766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/06/2023] [Accepted: 03/07/2023] [Indexed: 06/18/2023]
Abstract
Because of their exposure to air, eyes can come into contact with air pollutants such as particulate matter (PM), which may cause severe ocular pathologies. Prolonged ocular PM exposure may increase inflammation and endoplasmic reticulum stress in the retina. Herein, we investigated whether PM exposure induces ocular inflammation and endoplasmic reticulum (ER) stress-related cellular responses in human retinal epithelium-19 (ARPE-19) cells. To understand how PM promotes ocular inflammation, we monitored the activation of the mitogen-activated protein kinase (MAPK)/nuclear factor kappa beta (NFκB) axis and the expression of key inflammatory mRNAs. We also measured the upregulation of signature components for the ER-related unfolded protein response (UPR) pathways, as well as intracellular calcium ([Ca2+]i) levels, as readouts for ER stress induction following PM exposure. Ocular PM exposure significantly elevated the expression of multiple cytokine mRNAs and increased phosphorylation levels of NFκB-MAPK axis in a PM dose-dependent manner. Moreover, incubation with PM significantly increased [Ca2+]i levels and the expression of UPR-related proteins, which indicated ER stress resulting from cell hypoxia, and upregulation of hypoxic adaptation mechanisms such as the ER-associated UPR pathways. Our study demonstrated that ocular PM exposure increased inflammation in ARPE-19 cells, by activating the MAPK/NFκB axis and cytokine mRNA expression, while also inducing ER stress and stress adaptation responses. These findings may provide helpful insight into clinical and non-clinical research examining the role of PM exposure in ocular pathophysiology and delineating its underlying molecular mechanisms.
Collapse
Affiliation(s)
- Sunyoung Jeong
- Bioanalytical and Pharmacokinetic Research Group, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea
- Department of Human and Environmental Toxicology, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Eui-Cheol Shin
- Department of GreenBio Science/Food Science and Technology, Gyeongsang National University, Jinju 52725, Republic of Korea
| | - Jong-Hwa Lee
- Bioanalytical and Pharmacokinetic Research Group, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea
- Department of Human and Environmental Toxicology, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Jung-Heun Ha
- Department of Food Science and Nutrition, Dankook University, Cheonan 31116, Republic of Korea
- Research Center for Industrialization of Natural Neutralization, Dankook University, Yongin 16890, Republic of Korea
| |
Collapse
|
9
|
Lee S, Lee EJ, Lee GM, Yun JH, Yoo W. Inhibitory effect of fucoidan on TNF-α-induced inflammation in human retinal pigment epithelium cells. Front Nutr 2023; 10:1162934. [PMID: 37125026 PMCID: PMC10130517 DOI: 10.3389/fnut.2023.1162934] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 03/27/2023] [Indexed: 05/02/2023] Open
Abstract
Sargassum horneri (S. horneri) is a brown seaweed that contains a fucose-rich sulfated polysaccharide called fucoidan and is known to possess beneficial bioactivities, such as anti-inflammatory, antiviral, antioxidative, and antitumoral effects. This study aimed to determine the anti-inflammatory effects of AB_SH (hydrothermal extracts from S. horneri) and its bioactive compound (fucoidan) against tumor necrosis factor alpha (TNF-α)-induced inflammation in human retinal pigment epithelial (RPE) cells. AB_SH did not exhibit any cytotoxicity, and it decreased the mRNA expression of interleukin (IL)-6 and IL-8 and the production of the cytokines IL-6 and TNF-α. It also suppressed the expression levels of phosphorylated nuclear factor kappa B (NF-κB) and mitogen-activated protein kinases (MAPKs), including c-Jun amino-terminal kinases (JNK), p38 protein kinases (p38), and extracellular signal-regulated kinase (ERK) proteins, suggesting that AB_SH inhibits activation of the NF-kB/MAPK signaling pathway. Since fucoidan was identified in the composition analysis of AB_SH, it was additionally shown to be required for its anti-inflammatory effects in TNF-α-stimulated human RPE cells. In line with the AB_SH results, fucoidan reduced the mRNA levels of IL-6, IL-1ß, and IL-8 and production of the cytokines IL-6, TNF-α, and IL-8 through the downregulation of the NF-kB/MAPK signaling pathway in a dose-dependent manner. Collectively, the ability of AB_SH from S. horneri hydrothermal extracts to reduce inflammation indicates that it may be a good functional ingredient for managing ocular disorders.
Collapse
Affiliation(s)
- Sol Lee
- AceBiome Inc., Seoul, Republic of Korea
- R&D Center, AceBiome Inc., Daejeon, Republic of Korea
| | - Eun Jeoung Lee
- AceBiome Inc., Seoul, Republic of Korea
- R&D Center, AceBiome Inc., Daejeon, Republic of Korea
| | - Gyu Min Lee
- AceBiome Inc., Seoul, Republic of Korea
- R&D Center, AceBiome Inc., Daejeon, Republic of Korea
| | - Ji-Hyun Yun
- AceBiome Inc., Seoul, Republic of Korea
- R&D Center, AceBiome Inc., Daejeon, Republic of Korea
| | - Wonbeak Yoo
- AceBiome Inc., Seoul, Republic of Korea
- R&D Center, AceBiome Inc., Daejeon, Republic of Korea
- *Correspondence: Wonbeak Yoo,
| |
Collapse
|
10
|
Yang R, Liu Q, Zhang M. The Past and Present Lives of the Intraocular Transmembrane Protein CD36. Cells 2022; 12:cells12010171. [PMID: 36611964 PMCID: PMC9818597 DOI: 10.3390/cells12010171] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/19/2022] [Accepted: 12/27/2022] [Indexed: 01/03/2023] Open
Abstract
Cluster of differentiation 36 (CD36) belongs to the B2 receptors of the scavenger receptor class B family, which is comprised of single-chain secondary transmembrane glycoproteins. It is present in a variety of cell types, including monocytes, macrophages, microvascular endothelial cells, adipocytes, hepatocytes, platelets, skeletal muscle cells, kidney cells, cardiomyocytes, taste bud cells, and a variety of other cell types. CD36 can be localized on the cell surface, mitochondria, endoplasmic reticulum, and endosomes, playing a role in lipid accumulation, oxidative stress injury, apoptosis, and inflammatory signaling. Recent studies have found that CD36 is expressed in a variety of ocular cells, including retinal pigment epithelium (RPE), retinal microvascular endothelial cells, retinal ganglion cells (RGC), Müller cells, and photoreceptor cells, playing an important role in eye diseases, such as age-related macular degeneration (AMD), diabetic retinopathy (DR), and glaucoma. Therefore, a comprehensive understanding of CD36 function and downstream signaling pathways is of great significance for the prevention and treatment of eye diseases. This article reviews the molecular characteristics, distribution, and function of scavenger receptor CD36 and its role in ophthalmology in order to deepen the understanding of CD36 in eye diseases and provide new ideas for treatment strategies.
Collapse
Affiliation(s)
- Rucui Yang
- Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou 515041, China
- Department of Ophthalmology, Shantou University Medical College, Shantou University, Shantou 515041, China
| | - Qingping Liu
- Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou 515041, China
| | - Mingzhi Zhang
- Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou 515041, China
| |
Collapse
|
11
|
Galindez SM, Keightley A, Koulen P. Differential distribution of steroid hormone signaling networks in the human choroid-retinal pigment epithelial complex. BMC Ophthalmol 2022; 22:406. [PMID: 36266625 PMCID: PMC9583547 DOI: 10.1186/s12886-022-02585-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 08/22/2022] [Indexed: 11/10/2022] Open
Abstract
Background The retinal pigment epithelium (RPE), a layer of pigmented cells that lies between the neurosensory retina and the underlying choroid, plays a critical role in maintaining the functional integrity of photoreceptor cells and in mediating communication between the neurosensory retina and choroid. Prior studies have demonstrated neurotrophic effects of select steroids that mitigate the development and progression of retinal degenerative diseases via an array of distinct mechanisms of action. Methods Here, we identified major steroid hormone signaling pathways and their key functional protein constituents controlling steroid hormone signaling, which are potentially involved in the mitigation or propagation of retinal degenerative processes, from human proteome datasets with respect to their relative abundances in the retinal periphery, macula, and fovea. Results Androgen, glucocorticoid, and progesterone signaling networks were identified and displayed differential distribution patterns within these three anatomically distinct regions of the choroid-retinal pigment epithelial complex. Classical and non-classical estrogen and mineralocorticoid receptors were not identified. Conclusion Identified differential distribution patterns suggest both selective susceptibility to chronic neurodegenerative disease processes, as well as potential substrates for drug target discovery and novel drug development focused on steroid signaling pathways in the choroid-RPE.
Collapse
Affiliation(s)
- Sydney M Galindez
- School of Medicine, Vision Research Center, Department of Ophthalmology, University of Missouri - Kansas City School of Medicine, 2411 Holmes St, Kansas City, MO, 64108, USA
| | - Andrew Keightley
- School of Medicine, Vision Research Center, Department of Ophthalmology, University of Missouri - Kansas City School of Medicine, 2411 Holmes St, Kansas City, MO, 64108, USA
| | - Peter Koulen
- School of Medicine, Vision Research Center, Department of Ophthalmology, University of Missouri - Kansas City School of Medicine, 2411 Holmes St, Kansas City, MO, 64108, USA. .,Department of Biomedical Sciences, University of Missouri - Kansas City School of Medicine, Kansas City, MO, USA.
| |
Collapse
|
12
|
Immunological consequences of compromised ocular immune privilege accelerate retinal degeneration in retinitis pigmentosa. Orphanet J Rare Dis 2022; 17:378. [PMID: 36253797 PMCID: PMC9575261 DOI: 10.1186/s13023-022-02528-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 10/02/2022] [Indexed: 11/26/2022] Open
Abstract
Background Retinitis pigmentosa (RP) is a hereditary retinal disease which leads to visual impairment. The onset and progression of RP has physiological consequences that affects the ocular environment. Some of the key non-genetic factors which hasten the retinal degeneration in RP include oxidative stress, hypoxia and ocular inflammation. In this study, we investigated the status of the ocular immune privilege during retinal degeneration and the effect of ocular immune changes on the peripheral immune system in RP. We assessed the peripheral blood mononuclear cell stimulation by retinal antigens and their immune response status in RP patients. Subsequently, we examined alterations in ocular immune privilege machineries which may contribute to ocular inflammation and disease progression in rd1 mouse model. Results In RP patients, we observed a suppressed anti-inflammatory response to self-retinal antigens, thereby indicating a deviated response to self-antigens. The ocular milieu in rd1 mouse model indicated a significant decrease in immune suppressive ligands and cytokine TGF-B1, and higher pro-inflammatory ocular protein levels. Further, blood–retinal-barrier breakdown due to decrease in the expression of tight junction proteins was observed. The retinal breach potentiated pro-inflammatory peripheral immune activation against retinal antigens and caused infiltration of the peripheral immune cells into the ocular tissue. Conclusions Our studies with RP patients and rd1 mouse model suggest that immunological consequences in RP is a contributing factor in the progression of retinal degeneration. The ocular inflammation in the RP alters the ocular immune privilege mechanisms and peripheral immune response. These aberrations in turn create an auto-reactive immune environment and accelerate retinal degeneration.
Supplementary Information The online version contains supplementary material available at 10.1186/s13023-022-02528-x.
Collapse
|
13
|
Fernández-Vigo JI, Contreras I, Crespo MJ, Beckford C, Flores-Moreno I, Cobo-Soriano R, Pareja J, Martín MD, Moreno L, Arrevola-Velasco L. Expert Panel Consensus for Addressing Anti-VEGF Treatment Challenges of Diabetic Macular Edema in Spain. Clin Ophthalmol 2022; 16:3097-3106. [PMID: 36164581 PMCID: PMC9507974 DOI: 10.2147/opth.s374763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 08/03/2022] [Indexed: 11/23/2022] Open
Abstract
Purpose The treatment of diabetic macular edema (DME) has evolved rapidly in the past decade, highlighting the need to address the challenges of routine clinical practice decision-making through expert consensus agreements. Methods After a literature review and discussion of real-world experience on DME management, a group of ten retina specialists agreed on a consensus of recommendations for the most appropriate management of DME patients using vascular endothelial growth factor inhibitors (anti-VEGF) in Spain. Results The panel recommended early treatment initiation in DME patients with worse baseline visual acuity (VA) to maintain or improve outcome. For patients with good VA, an observation strategy was recommended, considering the presence of diabetic retinopathy, optical coherence tomography biomarkers, and impact on patient's quality of life. Based on the available evidence and clinical experience, the panel recommended the use of anti-VEGF intensive loading doses with the objective of achieving anatomic and visual responses as soon as possible, followed by a Treat & Extend (T&E) strategy to maintain VA improvement. Aflibercept was recommended for patients with a baseline decimal VA <0.5, followed by a T&E strategy, including the possibility to extend frequency of injections up to 16 weeks. Conclusion An expert panel proposes a consensus for the management of DME in Spain. Early treatment initiation with anti-VEGF in DME patients is recommended to maintain or improve VA; aflibercept is recommended for patients with a poor baseline VA.
Collapse
Affiliation(s)
- José Ignacio Fernández-Vigo
- Deparment of Ophthalmology, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid, Spain
| | - Inés Contreras
- Deparment of Ophthalmology, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigaciones Sanitarias (IRYCIS) and Clínica Rementería, Madrid, Spain
| | - María José Crespo
- Department of Ophthalmology, Hospital Universitario Infanta Leonor, Madrid, Spain
| | - Carlos Beckford
- Department of Ophthalmology, Hospital Universitario Príncipe de Asturias, Alcalá de Henares, Madrid, Spain
| | | | - Rosario Cobo-Soriano
- Department of Ophthalmology, Hospital Universitario Henares, Universidad Francisco de Vitoria, Madrid, Spain
| | - Jesús Pareja
- Department of Ophthalmology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | | | - Luis Moreno
- Department of Ophthalmology, Hospital 12 de Octubre, Madrid, Spain
| | - Luis Arrevola-Velasco
- Department of Ophthalmology, Clinica Baviera Instituto Oftalmológico Europeo, Madrid, Spain
| |
Collapse
|
14
|
Hottin C, Perron M, Roger JE. GSK3 Is a Central Player in Retinal Degenerative Diseases but a Challenging Therapeutic Target. Cells 2022; 11:cells11182898. [PMID: 36139472 PMCID: PMC9496697 DOI: 10.3390/cells11182898] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 09/12/2022] [Accepted: 09/13/2022] [Indexed: 11/24/2022] Open
Abstract
Glycogen synthase kinase 3 (GSK3) is a key regulator of many cellular signaling processes and performs a wide range of biological functions in the nervous system. Due to its central role in numerous cellular processes involved in cell degeneration, a rising number of studies have highlighted the interest in developing therapeutics targeting GSK3 to treat neurodegenerative diseases. Although recent works strongly suggest that inhibiting GSK3 might also be a promising therapeutic approach for retinal degenerative diseases, its full potential is still under-evaluated. In this review, we summarize the literature on the role of GSK3 on the main cellular functions reported as deregulated during retinal degeneration, such as glucose homeostasis which is critical for photoreceptor survival, or oxidative stress, a major component of retinal degeneration. We also discuss the interest in targeting GSK3 for its beneficial effects on inflammation, for reducing neovascularization that occurs in some retinal dystrophies, or for cell-based therapy by enhancing Müller glia cell proliferation in diseased retina. Together, although GSK3 inhibitors hold promise as therapeutic agents, we highlight the complexity of targeting such a multitasked kinase and the need to increase our knowledge of the impact of reducing GSK3 activity on these multiple cellular pathways and biological processes.
Collapse
Affiliation(s)
- Catherine Hottin
- Paris-Saclay Institute of Neuroscience, CERTO-Retina France, CNRS, Université Paris-Saclay, 91400 Saclay, France
| | - Muriel Perron
- Paris-Saclay Institute of Neuroscience, CERTO-Retina France, CNRS, Université Paris-Saclay, 91400 Saclay, France
| | - Jérôme E Roger
- Paris-Saclay Institute of Neuroscience, CERTO-Retina France, CNRS, Université Paris-Saclay, 91400 Saclay, France
| |
Collapse
|
15
|
Deletion of the Pedf gene leads to inflammation, photoreceptor loss and vascular disturbances in the retina. Exp Eye Res 2022; 222:109171. [PMID: 35809620 DOI: 10.1016/j.exer.2022.109171] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/25/2022] [Accepted: 06/30/2022] [Indexed: 11/24/2022]
Abstract
Retinal diseases are often accompanied by inflammation, vascular abnormalities, and neurodegeneration that decrease vision. Treatment with exogenous PEDF is widely shown to alleviate these conditions leading us to hypothesize that loss of function of the PEDF gene disrupts these pathways and leads to visual loss. Measurements were carried out by detailed phenotyping of PEDF null mice to assess expression of immunomodulators, glia activation, systemic inflammation, vascular disturbances, and visual sensitivity often associated with retinal pathologies. With a deletion of the Pedf gene, there was increased expression of several immune modulators in Pedf-/- retinas and serum with IL-2 and GM-CSF upregulated in both. Increases in retina glia activation and macrophage infiltration, levels of serum c-reactive protein (CRP), numbers of white and red blood cells and platelets and decreased blood glucose levels were all features associated with PEDF null mice. With PEDF gene deletion, there was also a notable increase in apoptosis in early developing retinas (PN3), reduced thickness of the photoreceptor layer, swelling of the inner plexiform layer, reduced retinal sensitivity and steady-state reduced activation of Erk and Akt, two signaling pathways used by PEDF. There is a substantial body of animal data emphasizing utility of PEDF treatment in homeostatic regulation of retinal diseases, including diabetic retinopathy and age-related macular degeneration but there is little agreement or evidence on the role of endogenous PEDF in retinal diseases. Our findings strongly support the concept that a deletion of the PEDF gene makes the retina vulnerable to diseases, and argue that endogenous PEDF plays a critical role in limiting pathological events in the retina.
Collapse
|
16
|
The correlation between serum albumin and diabetic retinopathy among people with type 2 diabetes mellitus: NHANES 2011–2020. PLoS One 2022; 17:e0270019. [PMID: 35709212 PMCID: PMC9202838 DOI: 10.1371/journal.pone.0270019] [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: 04/18/2022] [Accepted: 06/02/2022] [Indexed: 11/19/2022] Open
Abstract
Objectives The objective of this research aimed to investigate the correlation involving serum albumin with diabetic retinopathy (DR) in patients with type 2 diabetes mellitus (T2DM). Methods From 2011 to 2020, the National Health and Nutrition Examination Survey (NHANES) surveyed 45462 participants. We used the relevant data to conduct descriptive statistics, linear regression, and Logistic regression analysis. Results After adjusting for age, sex, and race, as well as all other variables, serum albumin was significantly negatively related to DR (P<0.001). Furthermore, after controlling for confounding factors, the third quartile (Q3) and the fourth quartile (Q4) had quite a negative significant relationship with the incidence of DR (P<0.01). The second quartile had a significant positive correlation with DR, whereas the observed negative correlations were not statistically meaningful (P>0.05). Conclusion Albumin levels in the serum have a quantitatively significant negative correlation with DR. Serum albumin levels in the blood can be used as a reference point for protracted follow-up of people with T2DM.
Collapse
|
17
|
Trotta MC, Gesualdo C, Petrillo F, Lepre CC, Della Corte A, Cavasso G, Maggiore G, Hermenean A, Simonelli F, D’Amico M, Rossi S. Resolution of Inflammation in Retinal Disorders: Briefly the State. Int J Mol Sci 2022; 23:4501. [PMID: 35562891 PMCID: PMC9100636 DOI: 10.3390/ijms23094501] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 04/13/2022] [Accepted: 04/16/2022] [Indexed: 12/24/2022] Open
Abstract
The most frequent retinal diseases, such as diabetic retinopathy, age-related macular degeneration and posterior uveitis, are underlined by oxidative stress or aging-induced retinal inflammation, which contributes to vision impairing or loss. Resolution of inflammation is emerging as a critical phase able to counteract the inflammatory process leading to the progression of retinal damage. Particularly, pro-resolving mediators (PMs) play a key role in the modulation of inflammatory exudates and could be considered a new target to be investigated in different inflammatory-autoimmune pathologies. Here, we highlight the most recent studies concerning the role of the main PMs (lipoxins, resolvins, prtectins, maresins and annexins) in retinal inflammation, in order to collect the best evidence in the field of inflammatory retinal damage resolution and to propose novel pharmacological approaches in the management of the most common retinal diseases.
Collapse
Affiliation(s)
- Maria Consiglia Trotta
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, Via Santa Maria di Costantinopoli 16, 80138 Naples, Italy; (M.C.T.); (F.P.); (C.C.L.); (M.D.)
| | - Carlo Gesualdo
- Eye Clinic, Multidisciplinary Department of Medical, Surgical and Dental Sciences, University of Campania “Luigi Vanvitelli”, Via Luigi De Crecchio 6, 80131 Naples, Italy; (C.G.); (A.D.C.); (G.C.); (F.S.)
| | - Francesco Petrillo
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, Via Santa Maria di Costantinopoli 16, 80138 Naples, Italy; (M.C.T.); (F.P.); (C.C.L.); (M.D.)
| | - Caterina Claudia Lepre
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, Via Santa Maria di Costantinopoli 16, 80138 Naples, Italy; (M.C.T.); (F.P.); (C.C.L.); (M.D.)
| | - Alberto Della Corte
- Eye Clinic, Multidisciplinary Department of Medical, Surgical and Dental Sciences, University of Campania “Luigi Vanvitelli”, Via Luigi De Crecchio 6, 80131 Naples, Italy; (C.G.); (A.D.C.); (G.C.); (F.S.)
| | - Giancuomo Cavasso
- Eye Clinic, Multidisciplinary Department of Medical, Surgical and Dental Sciences, University of Campania “Luigi Vanvitelli”, Via Luigi De Crecchio 6, 80131 Naples, Italy; (C.G.); (A.D.C.); (G.C.); (F.S.)
| | - Giulia Maggiore
- Department of Ophthalmology, University of Foggia, Viale Luigi Pinto 1, 71122 Foggia, Italy;
| | - Anca Hermenean
- “Aurel Ardelean” Institute of Life Sciences, Vasile Goldis Western University of Arad, 86 Revolutiei Av., 310414 Arad, Romania;
| | - Francesca Simonelli
- Eye Clinic, Multidisciplinary Department of Medical, Surgical and Dental Sciences, University of Campania “Luigi Vanvitelli”, Via Luigi De Crecchio 6, 80131 Naples, Italy; (C.G.); (A.D.C.); (G.C.); (F.S.)
| | - Michele D’Amico
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, Via Santa Maria di Costantinopoli 16, 80138 Naples, Italy; (M.C.T.); (F.P.); (C.C.L.); (M.D.)
| | - Settimio Rossi
- Eye Clinic, Multidisciplinary Department of Medical, Surgical and Dental Sciences, University of Campania “Luigi Vanvitelli”, Via Luigi De Crecchio 6, 80131 Naples, Italy; (C.G.); (A.D.C.); (G.C.); (F.S.)
| |
Collapse
|
18
|
Usategui-Martín R, Puertas-Neyra K, Galindo-Cabello N, Hernández-Rodríguez LA, González-Pérez F, Rodríguez-Cabello JC, González-Sarmiento R, Pastor JC, Fernandez-Bueno I. Retinal Neuroprotective Effect of Mesenchymal Stem Cells Secretome Through Modulation of Oxidative Stress, Autophagy, and Programmed Cell Death. Invest Ophthalmol Vis Sci 2022; 63:27. [PMID: 35486068 PMCID: PMC9055551 DOI: 10.1167/iovs.63.4.27] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022] Open
Abstract
Purpose Degenerative mechanisms of retinal neurodegenerative diseases (RND) share common cellular and molecular signalization pathways. Curative treatment does not exist and cell-based therapy, through the paracrine properties of mesenchymal stem cells (MSC), is a potential unspecific treatment for RND. This study aimed to evaluate the neuroprotective capability of human bone marrow (bm) MSC secretome and its potential to modulate retinal responses to neurodegeneration. Methods An in vitro model of spontaneous retinal neurodegeneration was used to compare three days of monocultured neuroretina (NR), NR cocultured with bmMSC, and NR cultured with bmMSC secretome. We evaluated retinal morphology markers (Lectin peanut agglutinin, rhodopsin, protein kinase C α isoform, neuronal-specific nuclear protein, glial fibrillary acidic protein, TdT-mediated dUTP nick-end labeling, and vimentin) and proteins involved in apoptosis (apoptosis-inductor factor, caspase-3), necroptosis (MLKL), and autophagy (p62). Besides, we analyzed the relative mRNA expression through qPCR of genes involved in apoptosis (BAX, BCL2, CASP3, CASP8, CASP9), necroptosis (MLKL, RIPK1, RIPK3), autophagy (ATG7, BCLIN1, LC3B, mTOR, SQSTM1), oxidative stress (COX2, CYBA, CYBB, GPX6, SOD1, TXN2, TXNRD1) and inflammation (IL1, IL6, IL10, TGFb1, TNFa). Results The bmMSC secretome preserves retinal morphology, limits pro-apoptotic- and pro-necroptotic-related gene and protein expression, modulates autophagy-related genes and proteins, and stimulates the activation of antioxidant-associated genes. Conclusions The neuroprotective ability of the bmMSC secretome is associated with activation of antioxidant machinery, modulation of autophagy, and inhibition of apoptosis and necroptosis during retinal degeneration. The neuroprotective effect of bmMSC secretomes in the presence/absence of MSC looks similar. Our current results reinforce the hypothesis that the human bmMSC secretome slows retinal neurodegeneration and may be a therapeutic option for treating RND.
Collapse
Affiliation(s)
- Ricardo Usategui-Martín
- Instituto Universitario de Oftalmobiología Aplicada, Retina Group, Universidad de Valladolid, Valladolid, Spain.,Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, Valladolid, Spain.,Red Temática de Investigación Cooperativa en Salud, Oftared, Instituto de Salud Carlos III, Valladolid, Spain.,RetiBrain (RED2018-102499-T), Ministerio de Ciencia, Innovación y Universidades, Valladolid, Spain
| | - Kevin Puertas-Neyra
- Instituto Universitario de Oftalmobiología Aplicada, Retina Group, Universidad de Valladolid, Valladolid, Spain
| | - Nadia Galindo-Cabello
- Instituto Universitario de Oftalmobiología Aplicada, Retina Group, Universidad de Valladolid, Valladolid, Spain.,Postgraduate Unit, Faculty of Biological Sciences, National University of San Marcos, Lima, Peru
| | | | - Fernando González-Pérez
- Group for Advanced Materials and Nanobiotechnology (GIR BIOFORGE), CIBER-BBN, Edificio LUCIA, Universidad de Valladolid, Paseo Belén 19, Valladolid, Spain
| | - José Carlos Rodríguez-Cabello
- Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, Valladolid, Spain.,Group for Advanced Materials and Nanobiotechnology (GIR BIOFORGE), CIBER-BBN, Edificio LUCIA, Universidad de Valladolid, Paseo Belén 19, Valladolid, Spain
| | - Rogelio González-Sarmiento
- Molecular Medicine Unit, Department of Medicine, University of Salamanca, Salamanca, Spain.,Institute of Biomedical Research of Salamanca, Salamanca, Spain.,Institute of Molecular and Cellular Biology of Cancer, University of Salamanca-CSIC, Salamanca, Spain
| | - José Carlos Pastor
- Instituto Universitario de Oftalmobiología Aplicada, Retina Group, Universidad de Valladolid, Valladolid, Spain.,Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, Valladolid, Spain.,Red Temática de Investigación Cooperativa en Salud, Oftared, Instituto de Salud Carlos III, Valladolid, Spain.,RetiBrain (RED2018-102499-T), Ministerio de Ciencia, Innovación y Universidades, Valladolid, Spain
| | - Ivan Fernandez-Bueno
- Instituto Universitario de Oftalmobiología Aplicada, Retina Group, Universidad de Valladolid, Valladolid, Spain.,Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, Valladolid, Spain.,Red Temática de Investigación Cooperativa en Salud, Oftared, Instituto de Salud Carlos III, Valladolid, Spain.,RetiBrain (RED2018-102499-T), Ministerio de Ciencia, Innovación y Universidades, Valladolid, Spain
| |
Collapse
|
19
|
Heydari M, Zare M, Badie MR, Watson RR, Talebnejad MR, Afarid M. Crocin as a vision supplement. Clin Exp Optom 2022; 106:249-256. [PMID: 35231199 DOI: 10.1080/08164622.2022.2039554] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Crocin is a natural ingredient of saffron (Crocus sativus L.) flower that has shown potential for application as a supplement in eye health and preserving vision. Crocin has been examined for its potential to treat various eye diseases such as glaucoma, macular dystrophies, diabetic retinopathy, and age-related macular degeneration. This review briefly discusses the role of crocin in different eye diseases. The underlying pathophysiological pathways involved in the effect of crocin on ophthalmic diseases are also reviewed. Preclinical evidence shows the cytoprotective, antioxidative, anti-inflammatory, and blood-flow enhancing effects of crocin in retinal tissue. Crocin also affects the retinal pathologies by activating PI3K/Akt and inhibiting NF-κB signalling pathways. Clinical evidence suggests that crocin improves outcomes in patients with retinal degenerations, retinal dystrophies, and glaucoma. Overall, crocin can be suggested as a potential vision supplement in healthy populations and patients with eye diseases. However, more clinical studies with larger sample sizes and longer follow-up durations are needed to confirm the current evidence.
Collapse
Affiliation(s)
- Mojtaba Heydari
- Research Center for Traditional Medicine and History of Medicine, Department of Persian Medicine, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.,Poostchi Ophthalmology Research Center, Department of Ophthalmology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mousa Zare
- Poostchi Ophthalmology Research Center, Department of Ophthalmology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Reza Badie
- Poostchi Ophthalmology Research Center, Department of Ophthalmology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Mohammad Reza Talebnejad
- Poostchi Ophthalmology Research Center, Department of Ophthalmology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehrdad Afarid
- Poostchi Ophthalmology Research Center, Department of Ophthalmology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
20
|
Pöstyéni E, Ganczer A, Kovács-Valasek A, Gabriel R. Relevance of Peptide Homeostasis in Metabolic Retinal Degenerative Disorders: Curative Potential in Genetically Modified Mice. Front Pharmacol 2022; 12:808315. [PMID: 35095518 PMCID: PMC8793341 DOI: 10.3389/fphar.2021.808315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 12/20/2021] [Indexed: 11/19/2022] Open
Abstract
The mammalian retina contains approximately 30 neuropeptides that are synthetized by different neuronal cell populations, glia, and the pigmented epithelium. The presence of these neuropeptides leaves a mark on normal retinal molecular processes and physiology, and they are also crucial in fighting various pathologies (e.g., diabetic retinopathy, ischemia, age-related pathologies, glaucoma) because of their protective abilities. Retinal pathologies of different origin (metabolic, genetic) are extensively investigated by genetically manipulated in vivo mouse models that help us gain a better understanding of the molecular background of these pathomechanisms. These models offer opportunities to manipulate gene expression in different cell types to help reveal their roles in the preservation of retinal health or identify malfunction during diseases. In order to assess the current status of transgenic technologies available, we have conducted a literature survey focused on retinal disorders of metabolic origin, zooming in on the role of retinal neuropeptides in diabetic retinopathy and ischemia. First, we identified those neuropeptides that are most relevant to retinal pathologies in humans and the two clinically most relevant models, mice and rats. Then we continued our analysis with metabolic disorders, examining neuropeptide-related pathways leading to systemic or cellular damage and rescue. Last but not least, we reviewed the available literature on genetically modified mouse strains to understand how the manipulation of a single element of any given pathway (e.g., signal molecules, receptors, intracellular signaling pathways) could lead either to the worsening of disease conditions or, more frequently, to substantial improvements in retinal health. Most attention was given to studies which reported successful intervention against specific disorders. For these experiments, a detailed evaluation will be given and the possible role of converging intracellular pathways will be discussed. Using these converging intracellular pathways, curative effects of peptides could potentially be utilized in fighting metabolic retinal disorders.
Collapse
Affiliation(s)
- Etelka Pöstyéni
- Department of Experimental Zoology and Neurobiology, University of Pécs, Pécs, Hungary
| | - Alma Ganczer
- Department of Experimental Zoology and Neurobiology, University of Pécs, Pécs, Hungary.,János Szentágothai Research Centre, University of Pécs, Pécs, Hungary
| | - Andrea Kovács-Valasek
- Department of Experimental Zoology and Neurobiology, University of Pécs, Pécs, Hungary
| | - Robert Gabriel
- Department of Experimental Zoology and Neurobiology, University of Pécs, Pécs, Hungary.,János Szentágothai Research Centre, University of Pécs, Pécs, Hungary
| |
Collapse
|
21
|
Kaur G, Singh NK. The Role of Inflammation in Retinal Neurodegeneration and Degenerative Diseases. Int J Mol Sci 2021; 23:ijms23010386. [PMID: 35008812 PMCID: PMC8745623 DOI: 10.3390/ijms23010386] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/24/2021] [Accepted: 12/28/2021] [Indexed: 12/19/2022] Open
Abstract
Retinal neurodegeneration is predominantly reported as the apoptosis or impaired function of the photoreceptors. Retinal degeneration is a major causative factor of irreversible vision loss leading to blindness. In recent years, retinal degenerative diseases have been investigated and many genes and genetic defects have been elucidated by many of the causative factors. An enormous amount of research has been performed to determine the pathogenesis of retinal degenerative conditions and to formulate the treatment modalities that are the critical requirements in this current scenario. Encouraging results have been obtained using gene therapy. We provide a narrative review of the various studies performed to date on the role of inflammation in human retinal degenerative diseases such as age-related macular degeneration, inherited retinal dystrophies, retinitis pigmentosa, Stargardt macular dystrophy, and Leber congenital amaurosis. In addition, we have highlighted the pivotal role of various inflammatory mechanisms in the progress of retinal degeneration. This review also offers an assessment of various therapeutic approaches, including gene-therapies and stem-cell-based therapies, for degenerative retinal diseases.
Collapse
Affiliation(s)
- Geetika Kaur
- Integrative Biosciences Center, Wayne State University, Detroit, MI 48202, USA;
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI 48202, USA
| | - Nikhlesh K. Singh
- Integrative Biosciences Center, Wayne State University, Detroit, MI 48202, USA;
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI 48202, USA
- Correspondence:
| |
Collapse
|
22
|
Flores‐Bellver M, Mighty J, Aparicio‐Domingo S, Li KV, Shi C, Zhou J, Cobb H, McGrath P, Michelis G, Lenhart P, Bilousova G, Heissel S, Rudy MJ, Coughlan C, Goodspeed AE, Becerra SP, Redenti S, Canto‐Soler MV. Extracellular vesicles released by human retinal pigment epithelium mediate increased polarised secretion of drusen proteins in response to AMD stressors. J Extracell Vesicles 2021; 10:e12165. [PMID: 34750957 PMCID: PMC8575963 DOI: 10.1002/jev2.12165] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 10/06/2021] [Accepted: 10/25/2021] [Indexed: 12/22/2022] Open
Abstract
Age-related macular degeneration (AMD) is a leading cause of blindness worldwide. Drusen are key contributors to the etiology of AMD and the ability to modulate drusen biogenesis could lead to therapeutic strategies to slow or halt AMD progression. The mechanisms underlying drusen biogenesis, however, remain mostly unknown. Here we demonstrate that under homeostatic conditions extracellular vesicles (EVs) secreted by retinal pigment epithelium (RPE) cells are enriched in proteins associated with mechanisms involved in AMD pathophysiology, including oxidative stress, immune response, inflammation, complement system and drusen composition. Furthermore, we provide first evidence that drusen-associated proteins are released as cargo of extracellular vesicles secreted by RPE cells in a polarised apical:basal mode. Notably, drusen-associated proteins exhibited distinctive directional secretion modes in homeostatic conditions and, differential modulation of this directional secretion in response to AMD stressors. These observations underpin the existence of a finely-tuned mechanism regulating directional apical:basal sorting and secretion of drusen-associated proteins via EVs, and its modulation in response to mechanisms involved in AMD pathophysiology. Collectively, our results strongly support an active role of RPE-derived EVs as a key source of drusen proteins and important contributors to drusen development and growth.
Collapse
Affiliation(s)
- Miguel Flores‐Bellver
- CellSight Ocular Stem Cell and Regeneration ProgramDepartment of OphthalmologySue Anschutz‐Rodgers Eye CenterUniversity of Colorado, School of MedicineAuroraColoradoUSA
| | - Jason Mighty
- Lehman CollegeBronxNew YorkUSA
- Biology Doctoral ProgramThe Graduate School and University CenterCity University of New YorkNew YorkNew YorkUSA
| | - Silvia Aparicio‐Domingo
- CellSight Ocular Stem Cell and Regeneration ProgramDepartment of OphthalmologySue Anschutz‐Rodgers Eye CenterUniversity of Colorado, School of MedicineAuroraColoradoUSA
| | - Kang V. Li
- CellSight Ocular Stem Cell and Regeneration ProgramDepartment of OphthalmologySue Anschutz‐Rodgers Eye CenterUniversity of Colorado, School of MedicineAuroraColoradoUSA
| | - Cui Shi
- Lehman CollegeBronxNew YorkUSA
- Biology Doctoral ProgramThe Graduate School and University CenterCity University of New YorkNew YorkNew YorkUSA
| | | | - Hannah Cobb
- CellSight Ocular Stem Cell and Regeneration ProgramDepartment of OphthalmologySue Anschutz‐Rodgers Eye CenterUniversity of Colorado, School of MedicineAuroraColoradoUSA
| | - Patrick McGrath
- Department of DermatologyUniversity of Colorado School of MedicineAuroraColoradoUSA
| | - German Michelis
- Section of Protein Structure and FunctionNEINIHBethesdaMarylandUSA
| | - Patricia Lenhart
- CellSight Ocular Stem Cell and Regeneration ProgramDepartment of OphthalmologySue Anschutz‐Rodgers Eye CenterUniversity of Colorado, School of MedicineAuroraColoradoUSA
| | - Ganna Bilousova
- Department of DermatologyUniversity of Colorado School of MedicineAuroraColoradoUSA
- Charles C. Gates Center for Regenerative MedicineUniversity of Colorado School of MedicineAuroraColoradoUSA
- Linda Crnic Institute for Down SyndromeUniversity of Colorado School of MedicineAuroraColoradoUSA
| | - Søren Heissel
- Proteomics Resource CenterThe Rockefeller UniversityNew YorkNew YorkUSA
| | - Michael J. Rudy
- Department of NeurologyUniversity of Colorado School of MedicineAuroraColoradoUSA
| | - Christina Coughlan
- University of Colorado Alzheimer's and Cognition CenterDepartment of NeurologyLinda Crnic Institute for Down SyndromeUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | - Andrew E. Goodspeed
- Department of PharmacologyUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
- University of Colorado Cancer CenterUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | | | - Stephen Redenti
- Lehman CollegeBronxNew YorkUSA
- Biology Doctoral ProgramThe Graduate School and University CenterCity University of New YorkNew YorkNew YorkUSA
- Biochemistry Doctoral ProgramThe Graduate SchoolCity University of New YorkNew YorkNew YorkUSA
| | - M. Valeria Canto‐Soler
- CellSight Ocular Stem Cell and Regeneration ProgramDepartment of OphthalmologySue Anschutz‐Rodgers Eye CenterUniversity of Colorado, School of MedicineAuroraColoradoUSA
- Charles C. Gates Center for Regenerative MedicineUniversity of Colorado School of MedicineAuroraColoradoUSA
| |
Collapse
|
23
|
Abd Eldaiem MS, Ahmed SA, Elsaeid AA, Hassan AA, Ghoneim DF, Ibrahim AM. Light-Emitting Diode Laser Therapy for Hyperoxia-Induced Retinal Abnormalities. J Lasers Med Sci 2021; 12:e64. [DOI: 10.34172/jlms.2021.64] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Accepted: 06/27/2021] [Indexed: 01/04/2023]
Abstract
Introduction: Hyperoxygenation is linked to numerous effects in a variety of organ systems. It can cause tissue damage by generating reactive oxygen species (ROS), increasing oxidative stress, and inducing cell death by apoptosis. The present study aimed to evaluate the effects of low-level laser therapy on the retina in response to acute hyperoxia in animals. Methods: A total of 70 Wistar albino rats were evaluated in the present study: 10 rats were designated as a control group, and the rest were exposed to hyperoxia (O2 , 90%) for 3 days, 1 week, and 2 weeks (20 rats each). Each group was divided into two subgroups (n=10), one of which was designated as hyperoxia only. The other was treated with a 670 nm light-emitting diode laser (2 sessions/one week, ~ 9.0 J/cm2 ) in each eye. The animals were euthanized, and their retinas were dissected for analysis of protein content, sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE), total antioxidant capacity (TAC), hydrogen peroxide (H2 O2 ), malondialdehyde (MDA), and histological examination. Results: We found that two weeks of hyperoxia induced an increase in retinal protein content (P<0.001), an alteration in the intensities and molecular weights of protein fractions, a significant decrease in the TAC level (P<0.01), and a noticeable increase in H2 O2 and MDA levels (P<0.001). Histological examination revealed fragmentation of the photoreceptors and neovascularization in the outer and inner plexiform layers. Furthermore, the data showed remarkable improvement in the retinal protein contents, oxidative state, and retinal structure after light-emitting diode laser therapy. Conclusion: Light-emitting diode laser therapy was found to be a useful treatment paradigm for reducing hyperoxia-induced retinal damage.
Collapse
Affiliation(s)
| | - Salwa Abdelkawi Ahmed
- Biophysics and Laser Science Unit, Vision Sciences Department, Research Institute of Ophthalmology, Giza, Egypt
| | | | - Aziza Ahmed Hassan
- Ophthalmic Unit, National Institute of Laser enhanced Science, Cairo University, Cairo, Egypt
| | - Dina Fouad Ghoneim
- Ophthalmic Unit, National Institute of Laser enhanced Science, Cairo University, Cairo, Egypt
| | | |
Collapse
|
24
|
Uemura A, Fruttiger M, D'Amore PA, De Falco S, Joussen AM, Sennlaub F, Brunck LR, Johnson KT, Lambrou GN, Rittenhouse KD, Langmann T. VEGFR1 signaling in retinal angiogenesis and microinflammation. Prog Retin Eye Res 2021; 84:100954. [PMID: 33640465 PMCID: PMC8385046 DOI: 10.1016/j.preteyeres.2021.100954] [Citation(s) in RCA: 169] [Impact Index Per Article: 42.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 02/12/2021] [Accepted: 02/19/2021] [Indexed: 12/13/2022]
Abstract
Five vascular endothelial growth factor receptor (VEGFR) ligands (VEGF-A, -B, -C, -D, and placental growth factor [PlGF]) constitute the VEGF family. VEGF-A binds VEGF receptors 1 and 2 (VEGFR1/2), whereas VEGF-B and PlGF only bind VEGFR1. Although much research has been conducted on VEGFR2 to elucidate its key role in retinal diseases, recent efforts have shown the importance and involvement of VEGFR1 and its family of ligands in angiogenesis, vascular permeability, and microinflammatory cascades within the retina. Expression of VEGFR1 depends on the microenvironment, is differentially regulated under hypoxic and inflammatory conditions, and it has been detected in retinal and choroidal endothelial cells, pericytes, retinal and choroidal mononuclear phagocytes (including microglia), Müller cells, photoreceptor cells, and the retinal pigment epithelium. Whilst the VEGF-A decoy function of VEGFR1 is well established, consequences of its direct signaling are less clear. VEGFR1 activation can affect vascular permeability and induce macrophage and microglia production of proinflammatory and proangiogenic mediators. However the ability of the VEGFR1 ligands (VEGF-A, PlGF, and VEGF-B) to compete against each other for receptor binding and to heterodimerize complicates our understanding of the relative contribution of VEGFR1 signaling alone toward the pathologic processes seen in diabetic retinopathy, retinal vascular occlusions, retinopathy of prematurity, and age-related macular degeneration. Clinically, anti-VEGF drugs have proven transformational in these pathologies and their impact on modulation of VEGFR1 signaling is still an opportunity-rich field for further research.
Collapse
Affiliation(s)
- Akiyoshi Uemura
- Department of Retinal Vascular Biology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan.
| | - Marcus Fruttiger
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London, EC1V 9EL, UK.
| | - Patricia A D'Amore
- Schepens Eye Research Institute of Massachusetts Eye and Ear, 20 Staniford Street, Boston, MA, 02114, USA.
| | - Sandro De Falco
- Angiogenesis Laboratory, Institute of Genetics and Biophysics "Adriano Buzzati-Traverso", Via Pietro Castellino 111, 80131 Naples, Italy; ANBITION S.r.l., Via Manzoni 1, 80123, Naples, Italy.
| | - Antonia M Joussen
- Department of Ophthalmology, Charité-Universitätsmedizin Berlin, Hindenburgdamm 30, 12200 Berlin, and Augustenburger Platz 1, 13353, Berlin, Germany.
| | - Florian Sennlaub
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012, Paris, France.
| | - Lynne R Brunck
- Bayer Consumer Care AG, Pharmaceuticals, Peter-Merian-Strasse 84, CH-4052 Basel, Switzerland.
| | - Kristian T Johnson
- Bayer Consumer Care AG, Pharmaceuticals, Peter-Merian-Strasse 84, CH-4052 Basel, Switzerland.
| | - George N Lambrou
- Bayer Consumer Care AG, Pharmaceuticals, Peter-Merian-Strasse 84, CH-4052 Basel, Switzerland.
| | - Kay D Rittenhouse
- Bayer Consumer Care AG, Pharmaceuticals, Peter-Merian-Strasse 84, CH-4052 Basel, Switzerland.
| | - Thomas Langmann
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Joseph-Stelzmann-Str. 9, 50931, Cologne, Germany.
| |
Collapse
|
25
|
Harsing LG, Szénási G, Zelles T, Köles L. Purinergic-Glycinergic Interaction in Neurodegenerative and Neuroinflammatory Disorders of the Retina. Int J Mol Sci 2021; 22:ijms22126209. [PMID: 34201404 PMCID: PMC8228622 DOI: 10.3390/ijms22126209] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/03/2021] [Accepted: 06/04/2021] [Indexed: 12/26/2022] Open
Abstract
Neurodegenerative–neuroinflammatory disorders of the retina seriously hamper human vision. In searching for key factors that contribute to the development of these pathologies, we considered potential interactions among purinergic neuromodulation, glycinergic neurotransmission, and microglia activity in the retina. Energy deprivation at cellular levels is mainly due to impaired blood circulation leading to increased release of ATP and adenosine as well as glutamate and glycine. Interactions between these modulators and neurotransmitters are manifold. First, P2Y purinoceptor agonists facilitate reuptake of glycine by glycine transporter 1, while its inhibitors reduce reverse-mode operation; these events may lower extracellular glycine levels. The consequential changes in extracellular glycine concentration can lead to parallel changes in the activity of NR1/NR2B type NMDA receptors of which glycine is a mandatory agonist, and thereby may reduce neurodegenerative events in the retina. Second, P2Y purinoceptor agonists and glycine transporter 1 inhibitors may indirectly inhibit microglia activity by decreasing neuronal or glial glycine release in energy-compromised retina. These inhibitions may have a role in microglia activation, which is present during development and progression of neurodegenerative disorders such as glaucomatous and diabetic retinopathies and age-related macular degeneration or loss of retinal neurons caused by thromboembolic events. We have hypothesized that glycine transporter 1 inhibitors and P2Y purinoceptor agonists may have therapeutic importance in neurodegenerative–neuroinflammatory disorders of the retina by decreasing NR1/NR2B NMDA receptor activity and production and release of a series of proinflammatory cytokines from microglial cells.
Collapse
Affiliation(s)
- Laszlo G. Harsing
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary; (T.Z.); (L.K.)
- Correspondence: ; Tel.: +36-1-210-4416
| | - Gábor Szénási
- Institute of Translational Medicine, Semmelweis University, H-1089 Budapest, Hungary;
| | - Tibor Zelles
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary; (T.Z.); (L.K.)
- Department of Oral Biology, Semmelweis University, H-1089 Budapest, Hungary
| | - László Köles
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary; (T.Z.); (L.K.)
- Department of Oral Biology, Semmelweis University, H-1089 Budapest, Hungary
| |
Collapse
|
26
|
Jadeja RN, Martin PM. Oxidative Stress and Inflammation in Retinal Degeneration. Antioxidants (Basel) 2021; 10:antiox10050790. [PMID: 34067655 PMCID: PMC8156590 DOI: 10.3390/antiox10050790] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 05/06/2021] [Indexed: 12/15/2022] Open
Affiliation(s)
- Ravirajsinh N. Jadeja
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA;
| | - Pamela M. Martin
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA;
- Department of Ophthalmology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
- James and Jean Culver Vision Discovery Institute, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
- Correspondence: ; Tel.: +70-6721-4220; Fax: +70-6721-6608
| |
Collapse
|
27
|
Appelbaum T, Murgiano L, Becker D, Santana E, Aguirre GD. Candidate Genetic Modifiers for RPGR Retinal Degeneration. Invest Ophthalmol Vis Sci 2021; 61:20. [PMID: 33326016 PMCID: PMC7745631 DOI: 10.1167/iovs.61.14.20] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Purpose To define genetic variants associated with variable severity of X-linked progressive retinal atrophy 1 (XLPRA1) caused by a five-nucleotide deletion in canine RPGR exon ORF15. Methods A genome-wide association study (GWAS) was performed in XLPRA1 phenotype informative pedigree. Whole genome sequencing (WGS) was used for mutational analysis of genes within the candidate genomic region. Retinas of normal and mutant dogs were used for gene expression, gene structure, and RNA duplex analyses. Results GWAS followed by haplotype phasing identified an approximately 4.6 Mb candidate genomic interval on CFA31 containing seven protein-coding genes expressed in retina (ROBO1, ROBO2, RBM11, NRIP1, HSPA13, SAMSN1, and USP25). Furthermore, we identified and characterized two novel lncRNAs, ROBO1-AS and ROBO2-AS, that display overlapping gene organization with axon guidance pathway genes ROBO1 and ROBO2, respectively, producing sense-antisense gene pairs. Notably, ROBO1-AS and ROBO2-AS act in cis to form lncRNA/mRNA duplexes with ROBO1 and ROBO2, respectively, suggesting important roles for these lncRNAs in the ROBO regulatory network. A subsequent WGS identified candidate genes within the genomic region on CFA31 that might be implicated in modifying severity of XLPRA1. This approach led to discovery of genetic variants in ROBO1, ROBO1-AS, ROBO2-AS, and USP25 that are strongly associated with the XLPRA1 moderate phenotype. Conclusions The study provides new insights into the genetic basis of phenotypic variation in severity of RPGRorf15-associated retinal degeneration. Our findings suggest an important role for ROBO pathways in disease progression further expanding on our previously reported changes of ROBO1 expression in XLPRA1 retinas.
Collapse
Affiliation(s)
- Tatyana Appelbaum
- Department of Clinical Sciences & Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Leonardo Murgiano
- Department of Clinical Sciences & Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Doreen Becker
- Department of Clinical Sciences & Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States.,Leibniz Institute for Farm Animal Biology (FBN), Institute of Genome Biology, Dummerstorf, Germany
| | - Evelyn Santana
- Department of Clinical Sciences & Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Gustavo D Aguirre
- Department of Clinical Sciences & Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| |
Collapse
|
28
|
Role of pyroptosis in diabetic retinopathy and its therapeutic implications. Eur J Pharmacol 2021; 904:174166. [PMID: 33979651 DOI: 10.1016/j.ejphar.2021.174166] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 05/05/2021] [Accepted: 05/05/2021] [Indexed: 12/16/2022]
Abstract
Pyroptosis has recently been established as a term of programmed-inflammatory cell death. Pyroptosis is mainly divided into two molecular signaling pathways, including caspase-1-dependent canonical and caspase-4/5/11-dependent non-canonical inflammasome pathways. Extensive investigations have reported inflammasome activation facilitates the maturation and secretion of the inflammatory factors interleukin-1β/18 (IL-1β/18), cleavage of gasdermin D (GSDMD), and leading to the stimulation of pyroptosis-mediated cell death. Furthermore, accumulating studies report NLRP3 inflammasome activation plays a significant role in triggering the pyroptosis-mediated cell death and promotes the pathogenesis of diabetic retinopathy (DR). Our current review elaborates on the molecular mechanisms of pyroptosis-signaling pathways and their potential roles in the pathogenesis and impact of DR development. We also emphasize several investigational molecules regulating key steps in pyroptotic-cell death to create new comprehensions and findings to explore the pathogenesis of DR advancement. Our narrative review concisely suggests these potential pharmacological agents could be promising therapies to treat and manage DR in the future.
Collapse
|
29
|
Gurley JM, Gmyrek GB, McClellan ME, Hargis EA, Hauck SM, Dozmorov MG, Wren JD, Carr DJJ, Elliott MH. Neuroretinal-Derived Caveolin-1 Promotes Endotoxin-Induced Inflammation in the Murine Retina. Invest Ophthalmol Vis Sci 2021; 61:19. [PMID: 33079993 PMCID: PMC7585394 DOI: 10.1167/iovs.61.12.19] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Purpose The immune-privileged environment and complex organization of retinal tissue support the retina's essential role in visual function, yet confound inquiries into cell-specific inflammatory effects that lead to dysfunction and degeneration. Caveolin-1 (Cav1) is an integral membrane protein expressed in several retinal cell types and is implicated in immune regulation. However, whether Cav1 promotes or inhibits inflammatory processes in the retina (as well as in other tissues) remains unclear. Previously, we showed that global-Cav1 depletion resulted in reduced retinal inflammatory cytokine production but paradoxically elevated retinal immune cell infiltration. We hypothesized that these disparate responses are the result of differential cell-specific Cav1 functions in the retina. Methods We used Cre/lox technology to deplete Cav1 specifically in the neural retinal (NR) compartment to clarify the role NR-specific Cav1 (NR-Cav1) in the retinal immune response to intravitreal inflammatory challenge induced by activation of Toll-like receptor-4 (TLR4). We used multiplex protein suspension array and flow cytometry to evaluate innate immune activation. Additionally, we used bioinformatics assessment of differentially expressed membrane-associated proteins to infer relationships between NR-Cav1 and immune response pathways. Results NR-Cav1 depletion, which primarily affects Müller glia Cav1 expression, significantly altered immune response pathway regulators, decreased retinal inflammatory cytokine production, and reduced retinal immune cell infiltration in response to LPS-stimulated inflammatory induction. Conclusions Cav1 expression in the NR compartment promotes the innate TLR4-mediated retinal tissue immune response. Additionally, we have identified novel potential immune modulators differentially expressed with NR-Cav1 depletion. This study further clarifies the role of NR-Cav1 in retinal inflammation.
Collapse
Affiliation(s)
- Jami M Gurley
- Department of Ophthalmology/Dean McGee Eye Institute, University of Oklahoma Health Sciences Center (OUHSC), Oklahoma City, Oklahoma, United States
| | - Grzegorz B Gmyrek
- Department of Ophthalmology/Dean McGee Eye Institute, University of Oklahoma Health Sciences Center (OUHSC), Oklahoma City, Oklahoma, United States
| | - Mark E McClellan
- Department of Ophthalmology/Dean McGee Eye Institute, University of Oklahoma Health Sciences Center (OUHSC), Oklahoma City, Oklahoma, United States
| | - Elizabeth A Hargis
- Department of Ophthalmology/Dean McGee Eye Institute, University of Oklahoma Health Sciences Center (OUHSC), Oklahoma City, Oklahoma, United States
| | - Stefanie M Hauck
- Research Unit Protein Science, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Munich, Germany
| | - Mikhail G Dozmorov
- Department of Biostatistics, Virginia Commonwealth University (VCU), Richmond, Virginia, United States
| | - Jonathan D Wren
- Arthritis and Clinical Immunology Research Program, Division of Genomics and Data Sciences, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, Oklahoma, United States
| | - Daniel J J Carr
- Department of Ophthalmology/Dean McGee Eye Institute, University of Oklahoma Health Sciences Center (OUHSC), Oklahoma City, Oklahoma, United States.,Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center (OUHSC), Oklahoma City, Oklahoma, United States
| | - Michael H Elliott
- Department of Ophthalmology/Dean McGee Eye Institute, University of Oklahoma Health Sciences Center (OUHSC), Oklahoma City, Oklahoma, United States
| |
Collapse
|
30
|
Scheive M, Yazdani S, Hajrasouliha AR. The utility and risks of therapeutic nanotechnology in the retina. Ther Adv Ophthalmol 2021; 13:25158414211003381. [PMID: 33817552 PMCID: PMC7989128 DOI: 10.1177/25158414211003381] [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: 05/16/2020] [Accepted: 02/23/2021] [Indexed: 01/06/2023] Open
Abstract
The clinical application of nanotechnology in medicine is promising for therapeutic, diagnostic, and surgical improvements in the near future. Nanotechnologies in nano-ophthalmology are in the early stages of application in clinical contexts, including ocular drug and gene delivery systems addressing eye disorders, particularly retinopathies. Retinal diseases are challenging to treat as current interventions, such as intravitreal injections, are limited by their invasive nature. This review examines nanotechnological approaches to retinal diseases in a clinical context. Nanotechnology has the potential to transform pharmacological and surgical interventions by overcoming limitations posed by the protective anatomical and physiological barriers that limit access to the retina. Preclinical research in the application of nanoparticles in diagnostics indicates that nanoparticles can enhance existing diagnostic and screening tools to detect diseases earlier and more easily and improve disease progression monitoring precision.
Collapse
Affiliation(s)
- Melanie Scheive
- Indiana University School of Medicine, Indianapolis, IN, USA
| | - Saeed Yazdani
- Indiana University-Purdue University Indianapolis, Indianapolis, IN, USA
| | - Amir R Hajrasouliha
- Assistant Professor of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Department of Ophthalmology, Indiana University School of Medicine, 1160 W Michigan St., Indianapolis, IN 46202, USA
| |
Collapse
|
31
|
Therapeutic Application of Exosomes in Inflammatory Diseases. Int J Mol Sci 2021; 22:ijms22031144. [PMID: 33498928 PMCID: PMC7865921 DOI: 10.3390/ijms22031144] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/20/2021] [Accepted: 01/22/2021] [Indexed: 02/07/2023] Open
Abstract
Immunomodulation is on the cusp of being an important therapy for treating many diseases, due to the significant role of the immune system in defending the human body. Although the immune system is an essential defense system, overactivity can result in diverse sicknesses such as inflammation and autoimmune disease. Exosomes are emerging as a state-of-the-art therapeutic strategy for treating an overactive immune system. Thus, in this review, we will thoroughly review therapeutic applications of exosomes in various inflammatory and autoimmune diseases. Finally, issues for an outlook to the future of exosomal therapy will be introduced.
Collapse
|
32
|
Dabouz R, Cheng CWH, Abram P, Omri S, Cagnone G, Sawmy KV, Joyal JS, Desjarlais M, Olson D, Weil AG, Lubell W, Rivera JC, Chemtob S. An allosteric interleukin-1 receptor modulator mitigates inflammation and photoreceptor toxicity in a model of retinal degeneration. J Neuroinflammation 2020; 17:359. [PMID: 33246504 PMCID: PMC7694438 DOI: 10.1186/s12974-020-02032-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 11/10/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Inflammation and particularly interleukin-1β (IL-1β), a pro-inflammatory cytokine highly secreted by activated immune cells during early AMD pathological events, contribute significantly to retinal neurodegeneration. Here, we identify specific cell types that generate IL-1β and harbor the IL-1 receptor (IL-1R) and pharmacologically validate IL-1β's contribution to neuro-retinal degeneration using the IL-1R allosteric modulator composed of the amino acid sequence rytvela (as well as the orthosteric antagonist, Kineret) in a model of blue light-induced retinal degeneration. METHODS Mice were exposed to blue light for 6 h and sacrificed 3 days later. Mice were intraperitoneally injected with rytvela, Kineret, or vehicle twice daily for 3 days. The inflammatory markers F4/80, NLRP3, caspase-1, and IL-1β were assessed in the retinas. Single-cell RNA sequencing was used to determine the cell-specific expression patterns of retinal Il1b and Il1r1. Macrophage-induced photoreceptor death was assessed ex vivo using retinal explants co-cultured with LPS-activated bone marrow-derived macrophages. Photoreceptor cell death was evaluated by the TUNEL assay. Retinal function was assessed by flash electroretinography. RESULTS Blue light markedly increased the mononuclear phagocyte recruitment and levels of inflammatory markers associated with photoreceptor death. Co-localization of NLRP3, caspase-1, and IL-1β with F4/80+ mononuclear phagocytes was clearly detected in the subretinal space, suggesting that these inflammatory cells are the main source of IL-1β. Single-cell RNA sequencing confirmed the immune-specific expression of Il1b and notably perivascular macrophages in light-challenged mice, while Il1r1 expression was found primarily in astrocytes, bipolar, and vascular cells. Retinal explants co-cultured with LPS/ATP-activated bone marrow-derived macrophages displayed a high number of TUNEL-positive photoreceptors, which was abrogated by rytvela treatment. IL-1R antagonism significantly mitigated the inflammatory response triggered in vivo by blue light exposure, and rytvela was superior to Kineret in preserving photoreceptor density and retinal function. CONCLUSION These findings substantiate the importance of IL-1β in neuro-retinal degeneration and revealed specific sources of Il1b from perivascular MPs, with its receptor Ilr1 being separately expressed on surrounding neuro-vascular and astroglial cells. They also validate the efficacy of rytvela-induced IL-1R modulation in suppressing detrimental inflammatory responses and preserving photoreceptor density and function in these conditions, reinforcing the rationale for clinical translation.
Collapse
Affiliation(s)
- Rabah Dabouz
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada.,Departments of Pediatrics, Ophthalmology, and Pharmacology, Hôpital Maisonneuve-Rosemont Research Center, 5415 Boul L'Assomption, Montreal, QC, H1T 2 M4, Canada.,Hôpital Sainte Justine Research Centre, Montreal, QC, Canada
| | - Colin W H Cheng
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada.,Departments of Pediatrics, Ophthalmology, and Pharmacology, Hôpital Maisonneuve-Rosemont Research Center, 5415 Boul L'Assomption, Montreal, QC, H1T 2 M4, Canada.,Hôpital Sainte Justine Research Centre, Montreal, QC, Canada
| | - Pénélope Abram
- Departments of Pediatrics, Ophthalmology, and Pharmacology, Hôpital Maisonneuve-Rosemont Research Center, 5415 Boul L'Assomption, Montreal, QC, H1T 2 M4, Canada
| | - Samy Omri
- Departments of Pediatrics, Ophthalmology, and Pharmacology, Hôpital Maisonneuve-Rosemont Research Center, 5415 Boul L'Assomption, Montreal, QC, H1T 2 M4, Canada
| | - Gael Cagnone
- Hôpital Sainte Justine Research Centre, Montreal, QC, Canada
| | | | | | - Michel Desjarlais
- Departments of Pediatrics, Ophthalmology, and Pharmacology, Hôpital Maisonneuve-Rosemont Research Center, 5415 Boul L'Assomption, Montreal, QC, H1T 2 M4, Canada
| | - David Olson
- Department of Obstetrics & Gynecology, University of Alberta, Edmonton, AB, Canada
| | - Alexander G Weil
- Department of Neurosurgery, Hôpital Sainte Justine, Montreal, QC, Canada
| | - William Lubell
- Department of Chemistry, University of Montreal, Montreal, QC, Canada
| | - José Carlos Rivera
- Departments of Pediatrics, Ophthalmology, and Pharmacology, Hôpital Maisonneuve-Rosemont Research Center, 5415 Boul L'Assomption, Montreal, QC, H1T 2 M4, Canada.,Hôpital Sainte Justine Research Centre, Montreal, QC, Canada
| | - Sylvain Chemtob
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada. .,Departments of Pediatrics, Ophthalmology, and Pharmacology, Hôpital Maisonneuve-Rosemont Research Center, 5415 Boul L'Assomption, Montreal, QC, H1T 2 M4, Canada. .,Hôpital Sainte Justine Research Centre, Montreal, QC, Canada.
| |
Collapse
|
33
|
Forshaw TRJ, Ahmed HJ, Kjær TW, Andréasson S, Sørensen TL. Full-field Electroretinography in Age-related Macular Degeneration: can retinal electrophysiology predict the subjective visual outcome of cataract surgery? Acta Ophthalmol 2020; 98:693-700. [PMID: 32275357 DOI: 10.1111/aos.14430] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 03/13/2020] [Indexed: 11/28/2022]
Abstract
PURPOSE Predicting the visual gain from cataract surgery when the main cause of vision loss is age-related macular degeneration may be difficult and warrants the need for an objective predictor of subjective outcome. Full-field electroretinography is an objective measure of overall retinal function. We therefore wanted to study if full-field electroretinography can predict subjective visual outcome using visual function questionnaire. METHODS Thirty-one patients with age-related macular degeneration operated for bilateral cataract underwent full-field electroretinography preoperatively. Full-field electroretinography was performed according to International Society for the Clinical Electrophysiology of Vision standards using a Ganzfeld bowl (RETI-port/scan 21, Roland, Berlin) and Dawson-Trick-Litzkow fibre electrodes. Vision-related quality of life was measured using the National Eye Institute Visual Function Questionnaire-39 before first-eye surgery and 4.12 ± 2.11 months after second-eye surgery. RESULTS Mean change in composite visual function questionnaire score after cataract surgery was 9.2 ± 11.9. The patients were divided into three groups: visual function questionnaire composite score increase >10 (n = 17); no change (n = 8); and decrease (n = 6). In the dark-adapted full-field electroretinography responses, we found a significant difference between the three groups in the 0.01 b-wave amplitude (p = 0.05), the 10.0 b-wave amplitude (p = 0.04) and a near-significant difference in 3.0 a-wave amplitude (p = 0.09). Other dark-adapted responses (the 3.0 b-wave and 10.0 a-wave) did not show any significant differences between the three groups, and neither did the light-adapted responses. CONCLUSION Patients with low dark-adapted responses on full-field electroretinography preoperatively experience a decrease in subjective vision-related quality of life, suggesting that maintained rod function before cataract surgery may be important.
Collapse
Affiliation(s)
- Thomas Richard Johansen Forshaw
- Department of Ophthalmology Zealand University Hospital Roskilde Denmark
- Faculty of Health and Medical Sciences University of Copenhagen Copenhagen Denmark
| | | | - Troels Wesenberg Kjær
- Faculty of Health and Medical Sciences University of Copenhagen Copenhagen Denmark
- Department of Neurophysiology Zealand University Hospital Roskilde Denmark
| | | | - Torben Lykke Sørensen
- Department of Ophthalmology Zealand University Hospital Roskilde Denmark
- Faculty of Health and Medical Sciences University of Copenhagen Copenhagen Denmark
| |
Collapse
|
34
|
Hong SC, Ha JH, Lee JK, Jung SH, Kim JC. In Vivo Anti-Inflammation Potential of Aster koraiensis Extract for Dry Eye Syndrome by the Protection of Ocular Surface. Nutrients 2020; 12:nu12113245. [PMID: 33113960 PMCID: PMC7690718 DOI: 10.3390/nu12113245] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 10/16/2020] [Accepted: 10/17/2020] [Indexed: 12/18/2022] Open
Abstract
Dry eye syndrome (DES) is a corneal disease often characterized by an irritating, itching feeling in the eyes and light sensitivity. Inflammation and endoplasmic reticulum (ER) stress may play a crucial role in the pathogenesis of DES, although the underlying mechanism remains elusive. Aster koraiensis has been used traditionally as an edible herb in Korea. It has been reported to have wound-healing and inhibitory effects against insulin resistance and inflammation. Here, we examined the inhibitory effects of inflammation and ER stress by A. koraiensis extract (AKE) in animal model and human retinal pigmented epithelial (ARPE-19) cells. Oral administration of AKE mitigated DE symptoms, including reduced corneal epithelial thickness, increased the gap between lacrimal gland tissues in experimental animals and decreased tear production. It also inhibited inflammatory responses in the corneal epithelium and lacrimal gland. Consequently, the activation of NF-κB was attenuated by the suppression of cyclooxygenase-1 (COX-1) and cyclooxygenase-2 (COX-2). Moreover, AKE treatment ameliorated TNF-α-inducible ocular inflammation and thapsigargin (Tg)-inducible ER stress in animal model and human retinal pigmented epithelial (ARPE-19) cells. These results prove that AKE prevents detrimental functional and histological remodeling on the ocular surface and in the lacrimal gland through inhibition of inflammation and ER stress, suggesting its potential as functional food material for improvement of DES.
Collapse
Affiliation(s)
- Sung-Chul Hong
- Natural Informatics Research Center, Korea Institute of Science and Technology (KIST), Gangneung 25451, Korea;
| | - Jung-Heun Ha
- Research Center for Industrialization of Natural Neutralization, Dankook University, Cheonan 31116, Korea;
- Department of Food Science and Nutrition, Dankook University, Cheonan 31116, Korea
| | - Jennifer K. Lee
- Food Science & Human Nutrition Department, University of Florida, Gainesville, FL 32611, USA;
| | - Sang Hoon Jung
- Natural Product Research Center, Korea Institute of Science and Technology (KIST), Gangneung 25451, Korea;
- Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology (UST), Daejeon 34113, Korea
| | - Jin-Chul Kim
- Natural Informatics Research Center, Korea Institute of Science and Technology (KIST), Gangneung 25451, Korea;
- Correspondence: ; Tel.: +82-33-650-3515
| |
Collapse
|
35
|
Lee D, Kang H, Yoon KY, Chang YY, Song HB. A mouse model of retinal hypoperfusion injury induced by unilateral common carotid artery occlusion. Exp Eye Res 2020; 201:108275. [PMID: 32991884 DOI: 10.1016/j.exer.2020.108275] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 09/25/2020] [Accepted: 09/25/2020] [Indexed: 12/12/2022]
Abstract
Retina, one of the highest oxygen demanding tissues, is vulnerable to vascular insufficiencies, and various ocular vascular disorders can cause chronic retinal ischemia. To investigate the pathophysiology, rodent models developed by bilateral common carotid artery occlusion (BCCAO) have been utilized. However, mice lack posterior communicating arteries in the circle of Willis and cannot endure the brain ischemia induced by the bilateral occlusion. A mouse model to better reflect the localized ischemic stress in the retina without affecting the brain is still needed. Here, we established a mouse model of ischemic injury by permanent unilateral common carotid artery occlusion (UCCAO). Adult male mice were subjected to UCCAO, and changes in the ipsilateral retina were examined in comparison with the contralateral retina. Delayed perfusion was observed in the ipsilateral retina right after the occlusion and was not recovered later on. Common features of retinal ischemia were observed: hypoxia-inducible factor (HIF) stabilization; upregulation of hypoxia-responsive genes; altered levels of cytokines and chemokines. Activation of astrocytes and Müller cells in the inner retina was detected at day 2, and thinning of the inner retinal layer became significant at week 10. Together, our model can simulate retinal ischemia with morphological and molecular changes. It can be utilized to investigate pathophysiology of ischemic retinopathies.
Collapse
Affiliation(s)
- Deokho Lee
- Department of Tropical Medicine and Parasitology and Institute of Endemic Diseases, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Heekyoung Kang
- Department of Tropical Medicine and Parasitology and Institute of Endemic Diseases, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Ki Young Yoon
- Department of Tropical Medicine and Parasitology and Institute of Endemic Diseases, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yuan Yi Chang
- Department of Tropical Medicine and Parasitology and Institute of Endemic Diseases, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hyun Beom Song
- Department of Tropical Medicine and Parasitology and Institute of Endemic Diseases, Seoul National University College of Medicine, Seoul, Republic of Korea; Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Republic of Korea; Department of Ophthalmology, Seoul National University Hospital, Seoul, Republic of Korea.
| |
Collapse
|
36
|
Mishra A, Mohan KV, Nagarajan P, Iyer S, Kesarwani A, Nath M, Moksha L, Bhattacharjee J, Das B, Jain K, Sahu P, Sinha P, Velapandian T, Upadhyay P. Peripheral blood-derived monocytes show neuronal properties and integration in immune-deficient rd1 mouse model upon phenotypic differentiation and induction with retinal growth factors. Stem Cell Res Ther 2020; 11:412. [PMID: 32967734 PMCID: PMC7510317 DOI: 10.1186/s13287-020-01925-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 08/17/2020] [Accepted: 09/04/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Cell therapy is one of the most promising therapeutic interventions for retinitis pigmentosa. In the current study, we aimed to assess if peripheral blood-derived monocytes which are highly abundant and accessible could be utilized as a potential candidate for phenotypic differentiation into neuron-like cells. METHODS The peripheral blood-derived monocytes were reconditioned phenotypically using extrinsic growth factors to induce pluripotency and proliferation. The reconditioned monocytes (RM) were further incubated with a cocktail of growth factors involved in retinal development and growth to induce retinal neuron-like properties. These cells, termed as retinal neuron-like cells (RNLCs) were characterized for their morphological, molecular and functional behaviour in vitro and in vivo. RESULTS The monocytes de-differentiated in vitro and acquired pluripotency with the expression of prominent stem cell markers. Treatment of RM with retinal growth factors led to an upregulation of neuronal and retinal lineage markers and downregulation of myeloid markers. These cells show morphological alterations resembling retinal neuron-like cells and expressed photoreceptor (PR) markers. The induced RNLCs also exhibited relative membrane potential change upon light exposure suggesting that they have gained some neuronal characteristics. Further studies showed that RNLCs could also integrate in an immune-deficient retinitis pigmentosa mouse model NOD.SCID-rd1 upon sub-retinal transplantation. The RNLCs engrafted in the inner nuclear layer (INL) and ganglion cell layer (GCL) of the RP afflicted retina. Mice transplanted with RNLCs showed improvement in depth perception, exploratory behaviour and the optokinetic response. CONCLUSIONS This proof-of-concept study demonstrates that reconditioned monocytes can be induced to acquire retinal neuron-like properties through differentiation using a defined growth media and can be a potential candidate for cell therapy-based interventions and disease modelling for ocular diseases.
Collapse
Affiliation(s)
- Alaknanda Mishra
- National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, 110067, India
| | - K Varsha Mohan
- National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, 110067, India
| | - Perumal Nagarajan
- National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, 110067, India
| | - Srikanth Iyer
- National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, 110067, India
| | - Ashwani Kesarwani
- National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, 110067, India
| | - Madhu Nath
- Department of Ocular Pharmacology, Dr. Rajendra Prasad Centre for Ophthalmic Sciences, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Laxmi Moksha
- Department of Ocular Pharmacology, Dr. Rajendra Prasad Centre for Ophthalmic Sciences, All India Institute of Medical Sciences, New Delhi, 110029, India
| | | | - Barun Das
- National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, 110067, India
| | - Kshama Jain
- National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, 110067, India
| | - Parul Sahu
- National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, 110067, India
| | - Prakriti Sinha
- National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, 110067, India
| | - T Velapandian
- Department of Ocular Pharmacology, Dr. Rajendra Prasad Centre for Ophthalmic Sciences, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Pramod Upadhyay
- National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, 110067, India.
| |
Collapse
|
37
|
Tauroursodeoxycholic Acid Protects Retinal Pigment Epithelial Cells from Oxidative Injury and Endoplasmic Reticulum Stress In Vitro. Biomedicines 2020; 8:biomedicines8090367. [PMID: 32967221 PMCID: PMC7555559 DOI: 10.3390/biomedicines8090367] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 09/10/2020] [Accepted: 09/15/2020] [Indexed: 12/12/2022] Open
Abstract
Retinal degeneration is characterized by the dysfunction of retinal cells. Oxidative and endoplasmic reticulum (ER) stress play an important role in the pathogenesis and progression of retinal degeneration. Tauroursodeoxycholic acid (TUDCA) has been demonstrated to have protective effects in in vitro and in vivo retinal degeneration models. To fully understand the molecular mechanisms of TUDCA’s protection, we first treated human retinal pigment epithelial (RPE) cells, ARPE-19, with H2O2 or H2O2 plus TUDCA for 24 h. RPE cells co-exposed to TUDCA had higher cell viability and lower cell death rate compared to cells exposed to H2O2 alone. TUDCA significantly increased antioxidant capacity in H2O2-treated RPE cells by decreasing the generation of reactive oxygen species (ROS) and Malondialdehyde (MDA), upregulating the expression of antioxidant genes, and increasing the generation of glutathione (GSH). TUDCA also inhibited inflammation in H2O2-challenged RPE cells by decreasing the expression of proinflammatory cytokines. Furthermore, TUDCA suppressed thapsigargin-induced ER stress in RPE cells, as demonstrated by decreased the expression of CCAAT-enhancer-binding protein homologous protein (CHOP) and apoptosis. Our present study suggests that TUDCA can protect RPE cells against oxidative damage, inflammation, and ER stress and may benefit patients with retinal degeneration.
Collapse
|
38
|
Lew DS, Mazzoni F, Finnemann SC. Microglia Inhibition Delays Retinal Degeneration Due to MerTK Phagocytosis Receptor Deficiency. Front Immunol 2020; 11:1463. [PMID: 32765507 PMCID: PMC7381113 DOI: 10.3389/fimmu.2020.01463] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 06/05/2020] [Indexed: 12/19/2022] Open
Abstract
Retinitis Pigmentosa (RP) is a group of inherited retinal diseases characterized by progressive loss of rod followed by cone photoreceptors. An especially early onset form of RP with blindness in teenage years is caused by mutations in mertk, the gene encoding the clearance phagocytosis receptor Mer tyrosine kinase (MerTK). The cause for blindness in mutant MerTK-associated RP (mutMerTK-RP) is the failure of retinal pigment epithelial cells in diurnal phagocytosis of spent photoreceptor outer segment debris. However, the early onset and very fast progression of degeneration in mutMerTK-RP remains unexplained. Here, we explored the role of microglia in the Royal College of Surgeons (RCS) rat model of mutMerTK-RP. We found elevated levels of inflammatory cytokines and CD68 microglia activation marker, and more ionized calcium-binding adapter molecule 1 (Iba-1) positive microglia in RCS retina when compared to wild-type retina as early as postnatal day 14 (P14). Strikingly, renewal of photoreceptor outer segments in P14 wild-type rat retina is still immature with low levels of RPE phagocytosis implying that at this early age lack of this process in RCS rats is unlikely to distress photoreceptors. Although the total number of Iba-1 positive retinal microglia remains constant from P14 to P30, we observed increasing numbers of microglia in the outer retina from P20 implying migration to the outer retina before onset of photoreceptor cell death at ~P25. Iba-1 and CD68 levels also increase in the retina during this time period suggesting microglia activation. To determine whether microglia affect the degenerative process, we suppressed retinal microglia in vivo using tamoxifen or a combination of tamoxifen and liposomal clodronate. Treatments partly prevented elevation of Iba-1 and CD68 and relocalization of microglia. Moreover, treatments led to partial but significant retention of photoreceptor viability and photoreceptor function. We conclude that loss of the phagocytosis receptor MerTK causes microglia activation and relocalization in the retina before lack of RPE phagocytosis causes overt retinal degeneration, and that microglia activities accelerate loss of photoreceptors in mutMerTK-RP. These results suggest that therapies targeting microglia may delay onset and slow the progression of this blinding disease.
Collapse
Affiliation(s)
- Deborah S Lew
- Department of Biological Sciences, Center for Cancer, Genetic Diseases and Gene Regulation, Fordham University, Bronx, NY, United States
| | - Francesca Mazzoni
- Department of Biological Sciences, Center for Cancer, Genetic Diseases and Gene Regulation, Fordham University, Bronx, NY, United States
| | - Silvia C Finnemann
- Department of Biological Sciences, Center for Cancer, Genetic Diseases and Gene Regulation, Fordham University, Bronx, NY, United States
| |
Collapse
|
39
|
Silva NJ, Nagashima M, Li J, Kakuk‐Atkins L, Ashrafzadeh M, Hyde DR, Hitchcock PF. Inflammation and matrix metalloproteinase 9 (Mmp-9) regulate photoreceptor regeneration in adult zebrafish. Glia 2020; 68:1445-1465. [PMID: 32034934 PMCID: PMC7317489 DOI: 10.1002/glia.23792] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 12/24/2019] [Accepted: 01/23/2020] [Indexed: 12/17/2022]
Abstract
Brain injury activates complex inflammatory signals in dying neurons, surviving neurons, and glia. Here, we establish that inflammation regulates the regeneration of photoreceptors in the zebrafish retina and determine the cellular expression and function of the inflammatory protease, matrix metalloproteinase 9 (Mmp-9), during this regenerative neurogenesis. Following photoreceptor ablation, anti-inflammatory treatment suppresses the number of injury-induced progenitors and regenerated photoreceptors. Upon photoreceptor injury, mmp-9 is induced in Müller glia and Müller glia-derived photoreceptor progenitors. Deleting mmp-9 results in over production of injury-induced progenitors and regenerated photoreceptors, but over time the absence of Mmp-9 compromises the survival of the regenerated cones. At all time-points studied, the levels of tnf-α are significantly elevated in mutant retinas. Anti-inflammatory treatment in mutants rescues the defects in cone survival. These data provide a link between injury-induced inflammation in the vertebrate CNS, Mmp-9 function during neuronal regeneration and the requirement of Mmp-9 for the survival of regenerated cones.
Collapse
Affiliation(s)
- Nicholas J. Silva
- Neuroscience Graduate ProgramUniversity of MichiganAnn ArborMichigan
- Department of Ophthalmology and Visual SciencesUniversity of MichiganAnn ArborMichigan
| | - Mikiko Nagashima
- Department of Ophthalmology and Visual SciencesUniversity of MichiganAnn ArborMichigan
| | - Jingling Li
- Department of Biological SciencesUniversity of Notre DameNotre DameIndiana
| | - Laura Kakuk‐Atkins
- Department of Ophthalmology and Visual SciencesUniversity of MichiganAnn ArborMichigan
| | - Milad Ashrafzadeh
- Department of Ophthalmology and Visual SciencesUniversity of MichiganAnn ArborMichigan
| | - David R. Hyde
- Department of Biological SciencesUniversity of Notre DameNotre DameIndiana
| | - Peter F. Hitchcock
- Neuroscience Graduate ProgramUniversity of MichiganAnn ArborMichigan
- Department of Ophthalmology and Visual SciencesUniversity of MichiganAnn ArborMichigan
| |
Collapse
|
40
|
Li J, Chen Y, Zhang X, Ye S, Yi J, Chen Q, Liu Q. Inhibition of acetylcholinesterase attenuated retinal inflammation via suppressing NF-κB activation. Exp Eye Res 2020; 195:108003. [PMID: 32184102 DOI: 10.1016/j.exer.2020.108003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 01/10/2020] [Accepted: 03/11/2020] [Indexed: 11/29/2022]
Abstract
Elevated inflammatory cytokines contribute to the pathogenesis of various retinal diseases such as diabetic retinopathy, retinal vasculitis and retinitis. However, the underlying mechanism of retinal inflammation remains largely unknown. Recent studies demonstrated that acetylcholinesterase (ACHE) is an inflammatory indicator in central neural system. This study was aimed to dissect the role of ACHE in retinal inflammation, and its mechanism of action. Retinal inflammation was induced by intravitreal injection of tumor necrosis factor-α (TNF-α) in heterozygous ACHE knockdown mice (ACHE+/-) and wild type mice (ACHE+/+). Donepezil, a well-known ACHE inhibitor, was administrated by daily gavage. Expression of ACHE and intercellular adherent molecule-1 (ICAM-1), infiltration of CD11b+ inflammatory cells, retinal leukostasis and vascular leakage was determined in both ACHE+/- and ACHE+/+ mice. ARPE-19 cells, a human retinal pigment epithelial cell line, were cultured for in vitro assay. Knockdown of ACHE was achieved by lipofectamine-mediated siRNA transfection and pharmaceutical suppression of ACHE was manipulated by donepezil. Cellular expression and distribution of ACHE, ICAM-1, and phosphorylation of NF-κB, IκB and IKKα/β were detected by western-blot analysis or immunocytochemistry. Retinal expression of ACHE was dramatically upregulated, in parallel with increased ICAM-1 expression, enhanced leukostasis and augmented CD11b+ inflammatory cell infiltration as well as vascular hyperpermeability in ACHE+/+ mice injected with TNF-α. However, TNF-α-injected ACHE+/- mice showed lower level of ICAM-1, less leukostasis and fewer infiltrated CD11b+ cells. Moreover, TNF-α-induced retinal vascular leakage was significantly reduced in ACHE+/- mice. Similarly, TNF-α-induced retinal inflammatory response were also attenuated by donepezil intervention. In addition, TNF-α treatment resulted in significant induction of ACHE, upregulation of ICAM-1 and nuclear translocation of NF-κB, phosphorylation of IκB and IKKα/β in ARPE-19 cells. However, inhibition of ACHE reduced TNF-α-induced phosphorylation of NF-κB, IκB and IKKα/β in ARPE-19 cells. The present study reveals a pivotal role of ACHE in retinal inflammation. Inhibition of ACHE attenuates retinal inflammation and retinal leakage likely through suppressing NF-κB signaling activation.
Collapse
Affiliation(s)
- Jingming Li
- Affiliated Eye Hospital of Nanchang University, Nanchang University, Nanchang, China.
| | - Yingying Chen
- Department of Ophthalmology, Hainan People's Hospital, Haikou, China
| | - Xian Zhang
- Affiliated Eye Hospital of Nanchang University, Nanchang University, Nanchang, China
| | - Sihao Ye
- Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Jinglin Yi
- Affiliated Eye Hospital of Nanchang University, Nanchang University, Nanchang, China
| | - Qian Chen
- Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China.
| | - Qiuping Liu
- Affiliated Eye Hospital of Nanchang University, Nanchang University, Nanchang, China.
| |
Collapse
|
41
|
Kalt W, Cassidy A, Howard LR, Krikorian R, Stull AJ, Tremblay F, Zamora-Ros R. Recent Research on the Health Benefits of Blueberries and Their Anthocyanins. Adv Nutr 2020; 11:224-236. [PMID: 31329250 PMCID: PMC7442370 DOI: 10.1093/advances/nmz065] [Citation(s) in RCA: 202] [Impact Index Per Article: 40.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Revised: 04/12/2019] [Accepted: 06/20/2019] [Indexed: 12/15/2022] Open
Abstract
Awareness of the human health benefits of blueberries is underpinned by a growing body of positive scientific evidence from human observational and clinical research, plus mechanistic research using animal and in vitro models. Blueberries contain a large number of phytochemicals, including abundant anthocyanin pigments. Of their various phytochemicals, anthocyanins probably make the greatest impact on blueberry health functionality. Epidemiological studies associate regular, moderate intake of blueberries and/or anthocyanins with reduced risk of cardiovascular disease, death, and type 2 diabetes, and with improved weight maintenance and neuroprotection. These findings are supported by biomarker-based evidence from human clinical studies. Among the more important healthful aspects of blueberries are their anti-inflammatory and antioxidant actions and their beneficial effects on vascular and glucoregulatory function. Blueberry phytochemicals may affect gastrointestinal microflora and contribute to host health. These aspects have implications in degenerative diseases and conditions as well as the aging process. More evidence, and particularly human clinical evidence, is needed to better understand the potential for anthocyanin-rich blueberries to benefit public health. However, it is widely agreed that the regular consumption of tasty, ripe blueberries can be unconditionally recommended.
Collapse
Affiliation(s)
- Wilhelmina Kalt
- Agriculture and Agri-Food Canada, Kentville Research and Development Centre, Kentville, Nova Scotia, Canada (retired)
| | - Aedin Cassidy
- Department of Nutrition, Norwich Medical School, University of East Anglia, Norwich, United Kingdom
| | - Luke R Howard
- Department of Food Science, University of Arkansas, Fayetteville, AR, USA
| | - Robert Krikorian
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati Academic Health Center, Cincinnati, OH, USA
| | - April J Stull
- Department of Human Ecology, University of Maryland Eastern Shore, Princess Anne, MD, USA
| | - Francois Tremblay
- Department of Ophthalmology and Visual Sciences and Department of Physiology and Biophysics, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Raul Zamora-Ros
- Unit of Nutrition and Cancer, Cancer Epidemiology Research Programme, Catalan Institute of Oncology, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
| |
Collapse
|
42
|
Chistyakov DV, Azbukina NV, Astakhova AA, Goriainov SV, Chistyakov VV, Tiulina VV, Baksheeva VE, Kotelin VI, Fedoseeva EV, Zamyatnin AA, Philippov PP, Kiseleva OA, Bessmertny AM, Senin II, Iomdina EN, Sergeeva MG, Zernii EY. Comparative lipidomic analysis of inflammatory mediators in the aqueous humor and tear fluid of humans and rabbits. Metabolomics 2020; 16:27. [PMID: 32052201 DOI: 10.1007/s11306-020-1650-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 02/04/2020] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Ocular inflammation is a key pathogenic factor in most blindness-causing visual disorders. It can manifest in the aqueous humor (AH) and tear fluid (TF) as alterations in polyunsaturated fatty acids (PUFAs) and their metabolites, oxylipins, lipid mediators, which are biosynthesized via enzymatic pathways involving lipoxygenase, cyclooxygenase or cytochrome P450 monooxygenase and specifically regulate inflammation and resolution pathways. OBJECTIVES This study aimed to establish the baseline patterns of PUFAs and oxylipins in AH and TF by their comprehensive lipidomic identification and profiling in humans in the absence of ocular inflammation and comparatively analyze these compounds in the eye liquids of rabbits, the species often employed in investigative ophthalmology. METHODS Ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) was used for qualitative and quantitative characterization of lipid compounds in the analyzed samples. RESULTS A total of 28 lipid compounds were identified, including phospholipid derivatives and PUFAs, as well as 22 oxylipins. Whereas the PUFAs included arachidonic, docosahexaenoic and eicosapentaenoic acids, the oxylipins were derived mainly from arachidonic, linoleic and α-linolenic acids. Remarkably, although the concentration of oxylipins in AH was lower compared to TF, these liquids showed pronounced similarity in their lipid profiles, which additionally exhibited noticeable interspecies concordance. CONCLUSION The revealed correlations confirm the feasibility of rabbit models for investigating pathogenesis and trialing therapies of human eye disorders. The identified metabolite patterns suggest enzymatic mechanisms of oxylipin generation in AH and TF and might be used as a reference in ocular inflammation studies.
Collapse
Affiliation(s)
- Dmitry V Chistyakov
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 1-40 Leninskye Gory, Moscow, Russia, 119992.
| | - Nadezhda V Azbukina
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 1-40 Leninskye Gory, Moscow, Russia, 119992
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia, 119234
| | - Alina A Astakhova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 1-40 Leninskye Gory, Moscow, Russia, 119992
| | | | | | - Veronika V Tiulina
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 1-40 Leninskye Gory, Moscow, Russia, 119992
| | - Viktoriia E Baksheeva
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 1-40 Leninskye Gory, Moscow, Russia, 119992
| | - Vladislav I Kotelin
- Helmholtz National Medical Research Center of Eye Diseases, Moscow, Russia, 105062
| | - Elena V Fedoseeva
- Helmholtz National Medical Research Center of Eye Diseases, Moscow, Russia, 105062
| | - Andrey A Zamyatnin
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 1-40 Leninskye Gory, Moscow, Russia, 119992
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russia, 119991
| | - Pavel P Philippov
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 1-40 Leninskye Gory, Moscow, Russia, 119992
| | - Olga A Kiseleva
- Helmholtz National Medical Research Center of Eye Diseases, Moscow, Russia, 105062
| | | | - Ivan I Senin
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 1-40 Leninskye Gory, Moscow, Russia, 119992
| | - Elena N Iomdina
- Helmholtz National Medical Research Center of Eye Diseases, Moscow, Russia, 105062
| | - Marina G Sergeeva
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 1-40 Leninskye Gory, Moscow, Russia, 119992
| | - Evgeni Yu Zernii
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 1-40 Leninskye Gory, Moscow, Russia, 119992.
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russia, 119991.
| |
Collapse
|
43
|
Mechanisms and Treatment of Light-Induced Retinal Degeneration-Associated Inflammation: Insights from Biochemical Profiling of the Aqueous Humor. Int J Mol Sci 2020; 21:ijms21030704. [PMID: 31973128 PMCID: PMC7038222 DOI: 10.3390/ijms21030704] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Accepted: 01/17/2020] [Indexed: 12/21/2022] Open
Abstract
Ocular inflammation contributes to the pathogenesis of blind-causing retinal degenerative diseases, such as age-related macular degeneration (AMD) or photic maculopathy. Here, we report on inflammatory mechanisms that are associated with retinal degeneration induced by bright visible light, which were revealed while using a rabbit model. Histologically and electrophysiologically noticeable degeneration of the retina is preceded and accompanied by oxidative stress and inflammation, as evidenced by granulocyte infiltration and edema in this tissue, as well as the upregulation of total protein, pro-inflammatory cytokines, and oxidative stress markers in aqueous humor (AH). Consistently, quantitative lipidomic studies of AH elucidated increase in the concentration of arachidonic (AA) and docosahexaenoic (DHA) acids and lyso-platelet activating factor (lyso-PAF), together with pronounced oxidative and inflammatory alterations in content of lipid mediators oxylipins. These alterations include long-term elevation of prostaglandins, which are synthesized from AA via cyclooxygenase-dependent pathways, as well as a short burst of linoleic acid derivatives that can be produced by both enzymatic and non-enzymatic free radical-dependent mechanisms. The upregulation of all oxylipins is inhibited by the premedication of the eyes while using mitochondria-targeted antioxidant SkQ1, whereas the accumulation of prostaglandins and lyso-PAF can be specifically suppressed by topical treatment with cyclooxygenase inhibitor Nepafenac. Interestingly, the most prominent antioxidant and anti-inflammatory benefits and overall retinal protective effects are achieved by simultaneous administrating of both drugs indicating their synergistic action. Taken together, these findings provide a rationale for using a combination of mitochondria-targeted antioxidant and cyclooxygenase inhibitor for the treatment of inflammatory components of retinal degenerative diseases.
Collapse
|
44
|
Mellal K, Omri S, Mulumba M, Tahiri H, Fortin C, Dorion MF, Pham H, Garcia Ramos Y, Zhang J, Pundir S, Joyal JS, Bouchard JF, Sennlaub F, Febbraio M, Hardy P, Gravel SP, Marleau S, Lubell WD, Chemtob S, Ong H. Immunometabolic modulation of retinal inflammation by CD36 ligand. Sci Rep 2019; 9:12903. [PMID: 31501473 PMCID: PMC6733801 DOI: 10.1038/s41598-019-49472-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 08/20/2019] [Indexed: 12/26/2022] Open
Abstract
In subretinal inflammation, activated mononuclear phagocytes (MP) play a key role in the progression of retinopathies. Little is known about the mechanism involved in the loss of photoreceptors leading to vision impairment. Studying retinal damage induced by photo-oxidative stress, we observed that cluster of differentiation 36 (CD36)-deficient mice featured less subretinal MP accumulation and attenuated photoreceptor degeneration. Moreover, treatment with a CD36-selective azapeptide ligand (MPE-001) reduced subretinal activated MP accumulation in wild type mice and preserved photoreceptor layers and function as assessed by electroretinography in a CD36-dependent manner. The azapeptide modulated the transcriptome of subretinal activated MP by reducing pro-inflammatory markers. In isolated MP, MPE-001 induced dissociation of the CD36-Toll-like receptor 2 (TLR2) oligomeric complex, decreasing nuclear factor-kappa B (NF-κB) and NLR family pyrin domain containing 3 (NLRP3) inflammasome activation. In addition, MPE-001 caused an aerobic metabolic shift in activated MP, involving peroxisome proliferator-activated receptor-γ (PPAR-γ) activation, which in turn mitigated inflammation. Accordingly, PPAR-γ inhibition blocked the cytoprotective effect of MPE-001 on photoreceptor apoptosis elicited by activated MP. By altering activated MP metabolism, MPE-001 decreased immune responses to alleviate subsequent inflammation-dependent neuronal injury characteristic of various vision-threatening retinal disorders.
Collapse
Affiliation(s)
- Katia Mellal
- Faculty of Pharmacy, Université de Montréal, Montreal, Canada
| | - Samy Omri
- Maisonneuve-Rosemont Hospital, Montréal, Canada
- Mperia Therapeutics, Montréal, Canada
| | | | - Houda Tahiri
- Departments of Pediatrics, Ophthalmology and Pharmacology, Université de Montréal, Montreal, Canada
| | - Carl Fortin
- Faculty of Pharmacy, Université de Montréal, Montreal, Canada
| | | | - Hung Pham
- Faculty of Pharmacy, Université de Montréal, Montreal, Canada
| | | | - Jinqiang Zhang
- Department of Chemistry, Université de Montréal, Montreal, Canada
| | - Sheetal Pundir
- Departments of Pediatrics, Ophthalmology and Pharmacology, Université de Montréal, Montreal, Canada
| | - Jean-Sébastien Joyal
- Departments of Pediatrics, Ophthalmology and Pharmacology, Université de Montréal, Montreal, Canada
| | - Jean-François Bouchard
- Neuropharmacology Laboratory, School of Optometry, Université de Montréal, Montreal, Canada
| | - Florian Sennlaub
- Institut de la Vision, Sorbonne Universités, INSERM, CNRS, Paris, France
| | - Maria Febbraio
- Department of Dentistry, University of Alberta, Edmonton, Canada
| | - Pierre Hardy
- Departments of Pediatrics, Ophthalmology and Pharmacology, Université de Montréal, Montreal, Canada
| | | | - Sylvie Marleau
- Faculty of Pharmacy, Université de Montréal, Montreal, Canada
| | - William D Lubell
- Department of Chemistry, Université de Montréal, Montreal, Canada
| | - Sylvain Chemtob
- Maisonneuve-Rosemont Hospital, Montréal, Canada.
- Departments of Pediatrics, Ophthalmology and Pharmacology, Université de Montréal, Montreal, Canada.
| | - Huy Ong
- Faculty of Pharmacy, Université de Montréal, Montreal, Canada.
| |
Collapse
|
45
|
Cheng SC, Huang WC, S Pang JH, Wu YH, Cheng CY. Quercetin Inhibits the Production of IL-1β-Induced Inflammatory Cytokines and Chemokines in ARPE-19 Cells via the MAPK and NF-κB Signaling Pathways. Int J Mol Sci 2019; 20:ijms20122957. [PMID: 31212975 PMCID: PMC6628093 DOI: 10.3390/ijms20122957] [Citation(s) in RCA: 141] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 06/08/2019] [Accepted: 06/13/2019] [Indexed: 12/27/2022] Open
Abstract
Quercetin, a bioflavonoid derived from vegetables and fruits, exerts anti-inflammatory effects in various diseases. Our previous study revealed that quercetin could suppress the expression of matrix metalloprotease-9 (MMP-9) and intercellular adhesion molecule-1 (ICAM-1) to achieve anti-inflammatory effects in tumor necrosis factor-α (TNF-α)-stimulated human retinal pigment epithelial (ARPE-19) cells. The present study explored whether quercetin can inhibit the interleukin-1β (IL-1β)-induced production of inflammatory cytokines and chemokines in ARPE-19 cells. Prior to stimulation by IL-1β, ARPE-19 cells were pretreated with quercetin at various concentrations (2.5–20 µM). The results showed that quercetin could dose-dependently decrease the mRNA and protein levels of ICAM-1, IL-6, IL-8 and monocyte chemoattractant protein-1 (MCP-1). It also attenuated the adherence of the human monocytic leukemia cell line THP-1 to IL-1β-stimulated ARPE-19 cells. We also demonstrated that quercetin inhibited signaling pathways related to the inflammatory process, including phosphorylation of mitogen-activated protein kinases (MAPKs), inhibitor of nuclear factor κ-B kinase (IKK)α/β, c-Jun, cAMP response element-binding protein (CREB), activating transcription factor 2 (ATF2) and nuclear factor (NF)-κB p65, and blocked the translocation of NF-κB p65 into the nucleus. Furthermore, MAPK inhibitors including an extracellular signal-regulated kinase (ERK) 1/2 inhibitor (U0126), a p38 inhibitor (SB202190) and a c-Jun N-terminal kinase (JNK) inhibitor (SP600125) decreased the expression of soluble ICAM-1 (sICAM-1), but not ICAM-1. U0126 and SB202190 could inhibit the expression of IL-6, IL-8 and MCP-1, but SP600125 could not. An NF-κB inhibitor (Bay 11-7082) also reduced the expression of ICAM-1, sICAM-1, IL-6, IL-8 and MCP-1. Taken together, these results provide evidence that quercetin protects ARPE-19 cells from the IL-1β-stimulated increase in ICAM-1, sICAM-1, IL-6, IL-8 and MCP-1 production by blocking the activation of MAPK and NF-κB signaling pathways to ameliorate the inflammatory response.
Collapse
Affiliation(s)
- Shu-Chen Cheng
- Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital, Taoyuan 33372, Taiwan.
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan.
| | - Wen-Chung Huang
- Graduate Institute of Health Industry Technology, Research Center for Chinese Herbal Medicine and Research Center for Food and Cosmetic Safety, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 33303, Taiwan.
- Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan.
| | - Jong-Hwei S Pang
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan.
- Department of Physical Medicine and Rehabilitation, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan.
| | - Yi-Hong Wu
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan.
- Division of Chinese Internal Medicine, Center for Traditional Chinese Medicine, Chang Gung Memorial Hospital, Taoyuan 33372, Taiwan.
- School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan.
| | - Ching-Yi Cheng
- Graduate Institute of Health Industry Technology, Research Center for Chinese Herbal Medicine and Research Center for Food and Cosmetic Safety, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 33303, Taiwan.
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Linkou 33305, Taiwan.
- Department of Ophthalmology, Chang Gung Memorial Hospital, Linkou 33305, Taiwan.
| |
Collapse
|
46
|
Bermúdez V, Tenconi PE, Giusto NM, Mateos MV. Lipopolysaccharide-Induced Autophagy Mediates Retinal Pigment Epithelium Cells Survival. Modulation by the Phospholipase D Pathway. Front Cell Neurosci 2019; 13:154. [PMID: 31327962 PMCID: PMC6497095 DOI: 10.3389/fncel.2019.00154] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 04/08/2019] [Indexed: 12/12/2022] Open
Abstract
Inflammation and oxidative stress are common factors involved in the pathogenesis of retinal diseases, such as aged-related macular degeneration (AMD) and diabetic retinopathy (DR). Autophagy is a catabolic process essential to cell survival in response to stress. This process is highly active in retinal pigment epithelium (RPE) cells. Our previous findings demonstrated that lipopolysaccharide (LPS) induces an inflammatory response of RPE cells that implies classical phospholipases D (PLD1 and 2) activation, cyclooxygenase-2 (COX-2) expression, prostaglandin E2 (PGE2) production and reduced cell viability. In this work, we studied the autophagic process and its modulation by the PLD pathway in D407 and ARPE-19 RPE cells exposed to LPS. LPS (10 μg/ml or 25 μg/ml) exposure for 24 h increased light chain 3B-II (LC3B-II) content (an autophagy marker) and LC3B-positive punctate structures in both RPE cell lines studied. Next, the drug bafilomycin A1 (BAF, 50 nM) was used to block the autophagic flux. In cells pre-incubated with BAF, LC3B-II and sequestosome 1 (SQSTM1/p62) levels and autophagosome-like structures were increased by LPS, demonstrating that the inflammatory injury increases the autophagic process in RPE cells. To study the role of the PLD pathway, cells were pre-incubated for 1 h with selective PLD1 (VU0359595) or PLD2 (VU0285655-1) inhibitors prior to LPS addition. Under control condition, LC3B-positive punctate structures were increased in cells pre-incubated with PLD2 inhibitor while with PLD1 inhibitor were increased in cells exposed to LPS. MTT reduction assays showed that early autophagy inhibitors, 3-methyladenin (3-MA) or LY294002, enhanced the loss in cell viability induced by LPS exposure for 48 h. On the contrary, the inhibition of PLD1 and PLD2 prevented the loss in cell viability induced by LPS. In conclusion, our results show that even though LPS treatment promotes an inflammatory response in RPE cells, it also triggers the activation of the autophagic process which in turn may serve as a protective mechanism for the cells. In addition, we demonstrate that the PLD pathway modulates the autophagic process in RPE cells. Our findings contribute to the knowledge of the molecular basis of retinal inflammatory and degenerative diseases and open new avenues for potential therapeutic exploration.
Collapse
Affiliation(s)
- Vicente Bermúdez
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Bahía Blanca, Argentina.,Departamento de Biología, Bioquímica y Farmacia (DBByF), Universidad Nacional del Sur (UNS), Bahía Blanca, Argentina
| | - Paula Estefanía Tenconi
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Bahía Blanca, Argentina.,Departamento de Biología, Bioquímica y Farmacia (DBByF), Universidad Nacional del Sur (UNS), Bahía Blanca, Argentina
| | - Norma María Giusto
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Bahía Blanca, Argentina.,Departamento de Biología, Bioquímica y Farmacia (DBByF), Universidad Nacional del Sur (UNS), Bahía Blanca, Argentina
| | - Melina Valeria Mateos
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Bahía Blanca, Argentina.,Departamento de Biología, Bioquímica y Farmacia (DBByF), Universidad Nacional del Sur (UNS), Bahía Blanca, Argentina
| |
Collapse
|
47
|
Homme RP, Singh M, Majumder A, George AK, Nair K, Sandhu HS, Tyagi N, Lominadze D, Tyagi SC. Remodeling of Retinal Architecture in Diabetic Retinopathy: Disruption of Ocular Physiology and Visual Functions by Inflammatory Gene Products and Pyroptosis. Front Physiol 2018; 9:1268. [PMID: 30233418 PMCID: PMC6134046 DOI: 10.3389/fphys.2018.01268] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 08/21/2018] [Indexed: 02/06/2023] Open
Abstract
Diabetic patients suffer from a host of physiological abnormalities beyond just those of glucose metabolism. These abnormalities often lead to systemic inflammation via modulation of several inflammation-related genes, their respective gene products, homocysteine metabolism, and pyroptosis. The very nature of this homeostatic disruption re-sets the overall physiology of diabetics via upregulation of immune responses, enhanced retinal neovascularization, upregulation of epigenetic events, and disturbances in cells' redox regulatory system. This altered pathophysiological milieu can lead to the development of diabetic retinopathy (DR), a debilitating vision-threatening eye condition with microvascular complications. DR is the most prevalent cause of irreversible blindness in the working-age adults throughout the world as it can lead to severe structural and functional remodeling of the retina, decreasing vision and thus diminishing the quality of life. In this manuscript, we attempt to summarize recent developments and new insights to explore the very nature of this intertwined crosstalk between components of the immune system and their metabolic orchestrations to elucidate the pathophysiology of DR. Understanding the multifaceted nature of the cellular and molecular factors that are involved in DR could reveal new targets for effective diagnostics, therapeutics, prognostics, preventive tools, and finally strategies to combat the development and progression of DR in susceptible subjects.
Collapse
Affiliation(s)
- Rubens P. Homme
- Eye and Vision Science Laboratory, Department of Physiology, University of Louisville School of Medicine, Louisville, KY, United States
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY, United States
| | - Mahavir Singh
- Eye and Vision Science Laboratory, Department of Physiology, University of Louisville School of Medicine, Louisville, KY, United States
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY, United States
| | - Avisek Majumder
- Eye and Vision Science Laboratory, Department of Physiology, University of Louisville School of Medicine, Louisville, KY, United States
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY, United States
| | - Akash K. George
- Eye and Vision Science Laboratory, Department of Physiology, University of Louisville School of Medicine, Louisville, KY, United States
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY, United States
| | - Kavya Nair
- Eye and Vision Science Laboratory, Department of Physiology, University of Louisville School of Medicine, Louisville, KY, United States
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY, United States
| | - Harpal S. Sandhu
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, Louisville, KY, United States
- Kentucky Lions Eye Center, University of Louisville School of Medicine, Louisville, KY, United States
| | - Neetu Tyagi
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY, United States
| | - David Lominadze
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY, United States
| | - Suresh C Tyagi
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY, United States
| |
Collapse
|
48
|
Arroba AI, Campos-Caro A, Aguilar-Diosdado M, Valverde ÁM. IGF-1, Inflammation and Retinal Degeneration: A Close Network. Front Aging Neurosci 2018; 10:203. [PMID: 30026694 PMCID: PMC6041402 DOI: 10.3389/fnagi.2018.00203] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 06/14/2018] [Indexed: 01/10/2023] Open
Abstract
Retinal degenerative diseases are a group of heterogeneous diseases that include age-related macular degeneration (AMD), retinitis pigmentosa (RP), and diabetic retinopathy (DR). The progressive degeneration of the retinal neurons results in a severe deterioration of the visual function. Neuroinflammation is an early hallmark of many neurodegenerative disorders of the retina including AMD, RP and DR. Microglial cells, key components of the retinal immune defense system, are activated in retinal degenerative diseases. In the microglia the interplay between the proinflammatory/classically activated or antiinflammatory/alternatively activated phenotypes is a complex dynamic process that occurs during the course of disease due to the different environmental signals related to pathophysiological conditions. In this regard, an adequate transition from the proinflammatory to the anti-inflammatory response is necessary to counteract retinal neurodegeneration and its subsequent damage that leads to the loss of visual function. Insulin like-growth factor-1 (IGF-1) has been considered as a pleiotropic factor in the retina under health or disease conditions and several effects of IGF-1 in retinal immune modulation have been described. In this review, we provide recent insights of inflammation as a common feature of retinal diseases (AMD, RP and RD) highlighting the role of microglia, exosomes and IGF-1 in this process.
Collapse
Affiliation(s)
- Ana I Arroba
- Alberto Sols Biomedical Research Institute (IIBm) (CSIC/UAM), Madrid, Spain.,Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERdem), ISCIII, Madrid, Spain.,Research Unit, Instituto de Investigación e Innovación en Ciencias Biomédicas de la Provincia de Cádiz (INiBICA), University Hospital "Puerta del Mar", Cádiz, Spain
| | - Antonio Campos-Caro
- Research Unit, Instituto de Investigación e Innovación en Ciencias Biomédicas de la Provincia de Cádiz (INiBICA), University Hospital "Puerta del Mar", Cádiz, Spain
| | - Manuel Aguilar-Diosdado
- Research Unit, Instituto de Investigación e Innovación en Ciencias Biomédicas de la Provincia de Cádiz (INiBICA), University Hospital "Puerta del Mar", Cádiz, Spain.,Department of Endocrinology and Metabolism, Instituto de Investigación e Innovación en Ciencias Biomédicas de la Provincia de Cádiz (INiBICA), University Hospital "Puerta del Mar", Cádiz, Spain
| | - Ángela M Valverde
- Alberto Sols Biomedical Research Institute (IIBm) (CSIC/UAM), Madrid, Spain.,Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERdem), ISCIII, Madrid, Spain
| |
Collapse
|
49
|
Distinct CD40L receptors mediate inflammasome activation and secretion of IL-1β and MCP-1 in cultured human retinal pigment epithelial cells. Exp Eye Res 2018; 170:29-39. [PMID: 29454857 DOI: 10.1016/j.exer.2018.02.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 02/14/2018] [Accepted: 02/14/2018] [Indexed: 11/22/2022]
Abstract
CD40L signaling occurs in several diseases with inflammatory components, including ocular and retinal diseases. However, it has never been evaluated as a pathogenic mechanism in age-related macular degeneration (AMD) or as an inducer of inflammasome formation in any cell type. mRNA and protein levels of CD40, IL-1β, NALP1, NALP3, caspase-1, and caspase-5 were determined by RT-PCR, qPCR, and Western blot. CD40L receptor (CD40, α5β1, and CD11b) expression was determined by Western and immunofluorescent staining. IL-1β, IL-18, and MCP-1 secretions were determined by ELISA. NALP1 and NALP3 inflammasome formation were determined by Co-IP. Experiments were conducted on primary human retinal pigment epithelial (hRPE) cells from four different donors. Human umbilical vein endothelial (HUVEC) and monocytic leukemia (THP-1) cells demonstrated the general applicability of our findings. In hRPE cells, CD40L-induced NALP1 and NALP3 inflammasome activation, cleavage of caspase-1 and caspase-5, and IL-1β and IL-18 secretion. Interestingly, neutralizing CD11b and α5β1 antibodies, but not CD40, reduced CD40L-induced IL-1β secretion in hRPE cells. Similarly, CD40L treatment also induced HUVEC and THP-1 cells to secret IL-1β through CD11b and α5β1. Additionally, the CD40L-induced IL-1β secretion acted in an autocrine/paracrine manner to feed back and induce hRPE cells to secrete MCP-1. This study is the first to show that CD40L induces inflammasome activation in any cell type, including hRPE cells, and that this induction is through CD11b and α5β1 cell-surface receptors. These mechanisms likely play an important role in many retinal and non-retinal diseases and provide compelling drug targets that may help reduce pro-inflammatory processes.
Collapse
|
50
|
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]
|