201
|
Shatz W, Aaronson J, Yohe S, Kelley RF, Kalia YN. Strategies for modifying drug residence time and ocular bioavailability to decrease treatment frequency for back of the eye diseases. Expert Opin Drug Deliv 2018; 16:43-57. [PMID: 30488721 DOI: 10.1080/17425247.2019.1553953] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
INTRODUCTION Treating posterior eye diseases has become a major area of focus for pharmaceutical and biotechnology companies. Current standard of care for treating posterior eye diseases relies on administration via intravitreal injection. Although effective, this is not without complications and there is great incentive to develop longer-acting therapeutics and/or sustained release delivery systems. Here, we present an overview of emerging technologies for delivery of biologics to the back of the eye. AREAS COVERED Posterior eye diseases, intravitreal injection, age-related macular degeneration, anti-VEGF, ocular pharmacokinetics, novel technologies to extend half-life, in vivo models, translation to the clinic, and hurdles to effective patient care. EXPERT OPINION Posterior eye diseases are a worldwide public health issue. Although anti-VEGF molecules represent a major advance for treating diseases involving choroidal neovascularization, frequent injection can be burdensome for patients and clinicians. There is a need for effective and patient-friendly treatments for posterior eye diseases. Many technologies that enable long-acting delivery to the back of the eye are being evaluated. However, successful development of novel therapies and delivery technologies is hampered by a multitude of factors, including patient education, translatability of in vitro/in vivo preclinical data to the clinic, and regulatory challenges associated with novel technologies.
Collapse
Affiliation(s)
- Whitney Shatz
- a Department of Protein Chemistry , Genentech , South San Francisco , CA , USA.,b School of Pharmaceutical Sciences , University of Geneva & University of Lausanne , Geneva , Switzerland
| | - Jeffrey Aaronson
- c Department of Drug Delivery , Genentech , South San Francisco , CA , USA
| | - Stefan Yohe
- c Department of Drug Delivery , Genentech , South San Francisco , CA , USA
| | - Robert F Kelley
- c Department of Drug Delivery , Genentech , South San Francisco , CA , USA
| | - Yogeshvar N Kalia
- b School of Pharmaceutical Sciences , University of Geneva & University of Lausanne , Geneva , Switzerland
| |
Collapse
|
202
|
Li XF, Li SY, Dai CM, Li JC, Huang DR, Wang JY. PP2A inhibition by LB-100 protects retinal pigment epithelium cells from UV radiation via activation of AMPK signaling. Biochem Biophys Res Commun 2018; 506:73-80. [PMID: 30340831 DOI: 10.1016/j.bbrc.2018.10.077] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 10/13/2018] [Indexed: 12/21/2022]
Abstract
AMP-activated protein kinase (AMPK) signaling activation can inhibit Ultra-violet (UV) radiation (UVR)-induced retinal pigment epithelium (RPE) cell injuries. LB-100 is a novel inhibitor of protein phosphatase 2A (PP2A), the AMPKα1 phosphatase. Here, our results demonstrated that LB-100 significantly inhibited UVR-induced viability reduction, cell death and apoptosis in established ARPE-19 cells and primary murine RPE cells. LB-100 activated AMPK, nicotinamide adenine dinucleotide phosphate (NADPH) and Nrf2 (NF-E2-related factor 2) signalings, inhibiting UVR-induced oxidative injuries and DNA damage in RPE cells. Conversely, AMPK inhibition, by AMPKα1-shRNA, -CRISPR/Cas9 knockout or -T172A mutation, almost blocked LB-100-induced RPE cytoprotection against UVR. Importantly, CRISPR/Cas9-mediated PP2A knockout mimicked and nullified LB-100-induced anti-UVR activity in RPE cells. Collectively, these results show that PP2A inhibition by LB-100 protects RPE cells from UVR via activation of AMPK signaling.
Collapse
Affiliation(s)
- Xiao-Feng Li
- Department of Ophthalmology, The Affiliated Huai'an NO.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Shu-Yan Li
- Department of Ophthalmology, The Affiliated Huai'an NO.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Chang-Ming Dai
- Department of Ophthalmology, The Affiliated Huai'an NO.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Jian-Chang Li
- Department of Ophthalmology, The Affiliated Huai'an NO.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Da-Rui Huang
- Department of Ophthalmology, The Affiliated Huai'an NO.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Jun-Ying Wang
- Department of ENT, The Affiliated Huai'an NO.1 People's Hospital of Nanjing Medical University, Huai'an, China.
| |
Collapse
|
203
|
Yang JW, Tseng ML, Fu YM, Kang CH, Cheng YT, Kuo PH, Tzeng CK, Chiou SH, Wu CY, Chen GY. Printable Graphene Oxide Micropatterns for a Bio-Subretinal Chip. Adv Healthc Mater 2018; 7:e1800365. [PMID: 30051620 DOI: 10.1002/adhm.201800365] [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: 04/08/2018] [Revised: 06/09/2018] [Indexed: 01/23/2023]
Abstract
Recently, implantable artificial subretinal chips using electronic components have replaced photoreceptors to serve as the most feasible treatment for retinal diseases. As such a chip that is meant to be implanted and used for very long periods, growing retinal cells on it to improve the electrical stimulation efficiency and attraction of neuronal elements remains a challenge. Here, an inkjet printing technology is employed to create graphene oxide (GO) micropatterns onto microelectrodes of a photovoltaic-powered implantable retinal chip. These GO micropatterns allow human retinal pigment epithelium (RPE) cells to specially attach and grow in each microelectrode. In addition, the cell proliferation, viability, and tight junction of RPE cells are improved during culturing. The development of a simple surface-coating technology would pave the way for the development of the first fully integrated and encapsulated retinal prostheses with biocompatible on-chip microelectrodes for long-term implantation, which could be effectively applied in retina tissue engineering and therapy.
Collapse
Affiliation(s)
- Jia-Wei Yang
- Department of Electrical and Computer Engineering; National Chiao Tung University; Hsinchu 300 Taiwan
- Institute of Biomedical Engineering; College of Electrical and Computer Engineering; National Chiao Tung University; Hsinchu 300 Taiwan
| | - Ming-Liang Tseng
- Institute of Biomedical Engineering; College of Electrical and Computer Engineering; National Chiao Tung University; Hsinchu 300 Taiwan
| | - Yu-Min Fu
- Microsystems Integration Laboratory; Department of Electronics Engineering; National Chiao Tung University; Hsinchu 300 Taiwan
| | - Che-Hao Kang
- Microsystems Integration Laboratory; Department of Electronics Engineering; National Chiao Tung University; Hsinchu 300 Taiwan
| | - Yu-Ting Cheng
- Microsystems Integration Laboratory; Department of Electronics Engineering; National Chiao Tung University; Hsinchu 300 Taiwan
| | - Po-Han Kuo
- Department of Electrical Engineering; National Chiao Tung University; Hsinchu 300 Taiwan
| | - Chi-Kuan Tzeng
- Department of Electrical Engineering; National Chiao Tung University; Hsinchu 300 Taiwan
| | - Shih-Hwa Chiou
- Institute of Pharmacology; School of Medicine; National Yang-Ming University; Taipei 112 Taiwan
- Department of Medical Research; Taipei Veterans General Hospital; Taipei 112 Taiwan
- Genomics Research Center; Academia Sinica; Taipei 115 Taiwan
| | - Chung-Yu Wu
- Department of Electrical Engineering; National Chiao Tung University; Hsinchu 300 Taiwan
| | - Guan-Yu Chen
- Institute of Biomedical Engineering; College of Electrical and Computer Engineering; National Chiao Tung University; Hsinchu 300 Taiwan
- Department of Biological Science and Technology; National Chiao Tung University; Hsinchu 300 Taiwan
- Institute of Biomedical Engineering; College of Electrical and Computer Engineering; National Chiao Tung University; Hsinchu 300 Taiwan
| |
Collapse
|
204
|
Spindler J, Zandi S, Pfister IB, Gerhardt C, Garweg JG. Cytokine profiles in the aqueous humor and serum of patients with dry and treated wet age-related macular degeneration. PLoS One 2018; 13:e0203337. [PMID: 30157273 PMCID: PMC6114931 DOI: 10.1371/journal.pone.0203337] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 08/17/2018] [Indexed: 02/07/2023] Open
Abstract
PURPOSE To identify disease-specific cytokine profile differences in the aqueous humor (AH) (other than the vascular endothelial growth factor) between patients with dry and treated wet age-related macular degeneration (AMD) and healthy controls. METHODS This retrospective study drew on a case-series of patients diagnosed with dry AMD (n = 25) and treated wet AMD (n = 19), as well as on healthy controls (no systemic therapy; n = 20) undergoing phacoemulsification or vitrectomy. Samples of AH and serum were collected in parallel at the beginning of surgery. The levels of 43 cytokines were simultaneously determined using the Bio-Plex® multiplex beads system. Differences between the three groups were statistically compared using the Kruskal-Wallis H-Test after applying the Bonferroni correction for multiple comparisons (p<0.0012). RESULTS The concentrations of three cytokines were elevated in the AH of patients with dry AMD (CXCL6; p = 0.00067) and treated wet AMD (CXCL5, CXCL6, MIG/XCXL; all p<0.001) relative to those in the healthy controls. No other differences between the three groups were identified. The AH levels of seven cytokines (16%), including CXCL6, ranged below the lower limit of quantitation of the assay. Without the correction for multiple comparisons (p<0.05), the levels of 31 of the 43 cytokines in the AH of patients with AMD would have differed significantly from those in the control. The systemic cytokine profiles (serum) were similar in all three groups. CONCLUSIONS No systematic differences in the AH cytokine environment were identified between patients with dry AMD and those with treated wet AMD. This finding might indicate that AMD is either the result of a persistent imbalance in the physiological tissue milieu, or that the localized process induces no significant change in the cytokine environment of the anterior ocular segment.
Collapse
Affiliation(s)
- Jan Spindler
- Swiss Eye Institute and Berner Augenklinik am Lindenhofspital, Bern, Switzerland
- University of Bern, Bern, Switzerland
| | - Souska Zandi
- Swiss Eye Institute and Berner Augenklinik am Lindenhofspital, Bern, Switzerland
| | - Isabel B. Pfister
- Swiss Eye Institute and Berner Augenklinik am Lindenhofspital, Bern, Switzerland
- University of Bern, Bern, Switzerland
| | - Christin Gerhardt
- Swiss Eye Institute and Berner Augenklinik am Lindenhofspital, Bern, Switzerland
- University of Bern, Bern, Switzerland
| | - Justus G. Garweg
- Swiss Eye Institute and Berner Augenklinik am Lindenhofspital, Bern, Switzerland
- University of Bern, Bern, Switzerland
| |
Collapse
|
205
|
Protective Effect of Melatonin against Oxidative Stress-Induced Apoptosis and Enhanced Autophagy in Human Retinal Pigment Epithelium Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:9015765. [PMID: 30174783 PMCID: PMC6098907 DOI: 10.1155/2018/9015765] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 06/01/2018] [Accepted: 06/06/2018] [Indexed: 12/20/2022]
Abstract
Age-related macular degeneration (AMD) affects the retinal macula and results in loss of vision, and AMD is the primary cause of blindness and severe visual impairment among elderly people worldwide. AMD is characterized by the accumulation of drusen in the Bruch's membrane and dysfunction of retinal pigment epithelial (RPE) cells and photoreceptors. The pathogenesis of AMD remains unclear, and no effective treatment exists. Accumulating evidence indicates that oxidative stress plays a critical role in RPE cell degeneration and AMD. Melatonin is an antioxidant that scavenges free radicals, and it has anti-inflammatory, antitumor, and antiangiogenic effects. This study investigated the antioxidative, antiapoptotic, and autophagic effects of melatonin on oxidative damage to RPE cells. We used hydrogen peroxide (H2O2) to stimulate reactive oxygen species production to cause cell apoptosis in ARPE-19 cell lines. Our findings revealed that treatment with melatonin significantly inhibited H2O2-induced RPE cell damage, decreased the apoptotic rate, increased the mitochondrial membrane potential, and increased the autophagy effect. Furthermore, melatonin reduced the Bax/Bcl-2 ratio and the expression levels of the apoptosis-associated proteins cytochrome c and caspase 7. Additionally, melatonin upregulated the expression of the autophagy-related proteins LC3-II and Beclin-1 and downregulated the expression of p62. Thus, melatonin's effects on autophagy and apoptosis can protect against H2O2-induced oxidative damage in human RPE cells. Melatonin may have multiple protective effects on human RPE cells against H2O2-induced oxidative damage.
Collapse
|
206
|
Lin YC, Horng LY, Sung HC, Wu RT. Sodium Iodate Disrupted the Mitochondrial-Lysosomal Axis in Cultured Retinal Pigment Epithelial Cells. J Ocul Pharmacol Ther 2018; 34:500-511. [PMID: 30020815 DOI: 10.1089/jop.2017.0073] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
PURPOSE Low doses of sodium iodate (NaIO3) impair visual function in experimental animals with selective damage to retinal pigment epithelium (RPE) and serve as a useful model to study diseases caused by RPE degeneration. Mitochondrial dysfunction and defective autophagy have been suggested to play important roles in normal aging as well as many neurodegenerative diseases. In this study, we examined whether NaIO3 treatment disrupted the mitochondrial-lysosomal axis in cultured RPE. METHODS The human RPE cell line, ARPE-19, was treated with low concentrations (≤500 μM) of NaIO3. The expression of proteins involved in the autophagic pathway and mitochondrial biogenesis was examined with Western blot. Intracellular acidic compartments and lipofuscinogenesis were evaluated by acridine orange staining and autofluorescence, respectively. Mitochondrial mass, mitochondrial membrane potential (MMP), and mitochondrial function were quantified by MitoTracker Green staining, tetramethylrhodamine methyl ester staining, and the MTT assay, respectively. Phagocytosis and the degradation of photoreceptor outer segments (POS) were assessed by fluorescence-based approaches and Western blot against rhodopsin. RESULTS Treatment with low concentrations of NaIO3 decreased cellular acidity, blocked autophagic flux, and resulted in increased lipofuscinogenesis in ARPE-19 cells. Despite increases in protein levels of Sirtuin 1 and PGC-1α, mitochondrial function was compromised, and this decrease was attributed to disrupted MMP. POS phagocytic activities decreased by 60% in NaIO3-treated cells, and the degradation of ingested POS was also impaired. Pretreatment and cotreatment with rapamycin partially rescued NaIO3-induced RPE dysfunction. CONCLUSIONS Low concentrations of NaIO3 disrupted the mitochondrial-lysosomal axis in RPE and led to impaired phagocytic activities and degradation capacities.
Collapse
Affiliation(s)
- Ying-Cheng Lin
- 1 Institute of Biopharmaceutical Sciences, School of Pharmaceutical Sciences, National Yang-Ming University , Taipei, Taiwan .,2 Department of Ophthalmology, Yang-Ming branch, Taipei City Hospital , Taipei, Taiwan
| | - Lin-Yea Horng
- 1 Institute of Biopharmaceutical Sciences, School of Pharmaceutical Sciences, National Yang-Ming University , Taipei, Taiwan .,3 Research Center for Drug Discovery, National Yang-Ming University , Taipei, Taiwan
| | - Hui-Ching Sung
- 3 Research Center for Drug Discovery, National Yang-Ming University , Taipei, Taiwan
| | - Rong-Tsun Wu
- 1 Institute of Biopharmaceutical Sciences, School of Pharmaceutical Sciences, National Yang-Ming University , Taipei, Taiwan .,3 Research Center for Drug Discovery, National Yang-Ming University , Taipei, Taiwan .,4 Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University , Kaohsiung, Taiwan
| |
Collapse
|
207
|
Li XM, Huang D, Yu Q, Yang J, Yao J. Neuroligin-3 protects retinal cells from H2O2-induced cell death via activation of Nrf2 signaling. Biochem Biophys Res Commun 2018; 502:166-172. [DOI: 10.1016/j.bbrc.2018.05.141] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 05/19/2018] [Indexed: 01/12/2023]
|
208
|
Li XF, Liu XM, Huang DR, Cao HJ, Wang JY. PF-06409577 activates AMPK signaling to protect retinal pigment epithelium cells from UV radiation. Biochem Biophys Res Commun 2018; 501:293-299. [DOI: 10.1016/j.bbrc.2018.05.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 05/02/2018] [Indexed: 01/01/2023]
|
209
|
Bhuachalla BN, McGarrigle CA, O'Leary N, Akuffo KO, Peto T, Beatty S, Kenny RA. Orthostatic hypertension as a risk factor for age-related macular degeneration: Evidence from the Irish longitudinal study on ageing. Exp Gerontol 2018; 106:80-87. [DOI: 10.1016/j.exger.2018.02.029] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 02/25/2018] [Indexed: 10/17/2022]
|
210
|
Hutton-Smith LA, Gaffney EA, Byrne HM, Caruso A, Maini PK, Mazer NA. Theoretical Insights into the Retinal Dynamics of Vascular Endothelial Growth Factor in Patients Treated with Ranibizumab, Based on an Ocular Pharmacokinetic/Pharmacodynamic Model. Mol Pharm 2018; 15:2770-2784. [PMID: 29734810 DOI: 10.1021/acs.molpharmaceut.8b00280] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Neovascular age-related macular degeneration (wet AMD) results from the pathological angiogenesis of choroidal capillaries, which leak fluid within or below the macular region of the retina. The current standard of care for treating wet AMD utilizes intravitreal injections of anti-VEGF antibodies or antibody fragments to suppress ocular vascular endothelial growth factor (VEGF) levels. While VEGF suppression has been demonstrated in wet AMD patients by serial measurements of free-VEGF concentrations in aqueous humor samples, it is presumed that anti-VEGF molecules also permeate across the inner limiting membrane (ILM) of the retina as well as the retinal pigmented epithelium (RPE) and suppress VEGF levels in the retina and/or choroidal regions. The latter effects are inferred from serial optical coherence tomography (OCT) measurements of fluid in the retinal and sub-retinal spaces. In order to gain theoretical insights to the dynamics of retinal levels of free-VEGF following intravitreal injection of anti-VEGF molecules, we have extended our previous two-compartment pharmacokinetic/pharmacodynamic (PK/PD) model of ranibizumab-VEGF suppression in vitreous and aqueous humors to a three-compartment model that includes the retinal compartment. In the new model, reference values for the macromolecular permeability coefficients between retina and vitreous ( pILM) and between retina and choroid ( pRPE) were estimated from PK data obtained in rabbit. With these values, the three-compartment model was used to re-analyze the aqueous humor levels of free-VEGF obtained in wet AMD patients treated with ranibizumab and to compare them to the simulated retinal levels of free-VEGF, including the observed variability in PK and PD. We have also used the model to explore the impact of varying pILM and pRPE to assess the case in which an anti-VEGF molecule is impermeable to the ILM and to assess the potential effects of AMD pathology on the RPE barrier. Our simulations show that, for the reference values of pILM and pRPE, the simulated duration of VEGF suppression in the retina is approximately 50% shorter than the observed duration of VEGF suppression in the aqueous humor, a finding that may explain the short duration of suppressed disease activity in the "high anti-VEGF demand" patients reported by Fauser and Muether ( Br. J. Ophthalmol. 2016, 100, 1494-1498 ). At 10-fold lower values of pRPE, the durations of VEGF suppression in the retina and aqueous humor are comparable. Lastly we have used the model to explore the impact of dose and binding parameters on the duration and depth of VEGF suppression in the aqueous and retinal compartments. Our simulations with the three-compartment PK/PD model provide new insights into inter-patient variability in response to anti-VEGF therapy and offer a mechanistic framework for developing treatment regimens and molecules that may prolong the duration of retinal VEGF suppression.
Collapse
Affiliation(s)
- Laurence A Hutton-Smith
- Wolfson Centre for Mathematical Biology, Mathematical Institute, Andrew Wiles Building , University of Oxford , Radcliffe Observatory Quarter, Woodstock Road , Oxford OX2 6GG , U.K
| | - Eamonn A Gaffney
- Wolfson Centre for Mathematical Biology, Mathematical Institute, Andrew Wiles Building , University of Oxford , Radcliffe Observatory Quarter, Woodstock Road , Oxford OX2 6GG , U.K
| | - Helen M Byrne
- Wolfson Centre for Mathematical Biology, Mathematical Institute, Andrew Wiles Building , University of Oxford , Radcliffe Observatory Quarter, Woodstock Road , Oxford OX2 6GG , U.K
| | - Antonello Caruso
- Roche Pharma Research and Early Development, Pharmaceutical Sciences , Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd , Grenzacherstrasse 124 , 4070 Basel , Switzerland
| | - Philip K Maini
- Wolfson Centre for Mathematical Biology, Mathematical Institute, Andrew Wiles Building , University of Oxford , Radcliffe Observatory Quarter, Woodstock Road , Oxford OX2 6GG , U.K
| | - Norman A Mazer
- Roche Pharma Research and Early Development, Pharmaceutical Sciences , Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd , Grenzacherstrasse 124 , 4070 Basel , Switzerland
| |
Collapse
|
211
|
ID2 protects retinal pigment epithelium cells from oxidative damage through p-ERK1/2/ID2/NRF2. Arch Biochem Biophys 2018; 650:1-13. [PMID: 29753724 DOI: 10.1016/j.abb.2018.05.008] [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: 02/07/2018] [Revised: 05/07/2018] [Accepted: 05/09/2018] [Indexed: 12/19/2022]
Abstract
Age-related macular degeneration (AMD) is the leading cause of blindness during aging. The degeneration of retinal pigment epithelium (RPE) is the main pathologic characteristic of AMD. ID2 is a member of the Inhibitor of DNA binding proteins (ID) family and is involved in regulation of cell proliferation and differentiation. However, currently the role of ID2 in oxidative injury response in RPE cells remains unknown. Here we showed that oxidative stress increased ID2 expression in RPE cells. Knockdown of ID2 promoted cell apoptosis and increased ROS level in RPE cells that were subjected to oxidative damage. In addition, over-expression of ID2 attenuated the oxidative damage response in RPE cells. Mechanistically, ID2 protected RPE cells from oxidative damage through activating NRF2. Furthermore, phosphorylation of ERK1/2 positively regulated the protective function of ID2. Finally, we confirmed that the oxidative damage increased Id2 expression and over-expression of Id2 elevated Nrf2 expression in primary mouse RPE cells. Therefore, ID2 protects RPE cells from oxidative damage through the p-ERK1/2/ID2/NRF2 pathway. Our study contributes to a better understanding of the mechanisms underlying oxidative stress in AMD and may present a new strategy for AMD treatment.
Collapse
|
212
|
Katschke KJ, Xi H, Cox C, Truong T, Malato Y, Lee WP, McKenzie B, Arceo R, Tao J, Rangell L, Reichelt M, Diehl L, Elstrott J, Weimer RM, van Lookeren Campagne M. Classical and alternative complement activation on photoreceptor outer segments drives monocyte-dependent retinal atrophy. Sci Rep 2018; 8:7348. [PMID: 29743491 PMCID: PMC5943270 DOI: 10.1038/s41598-018-25557-8] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 04/19/2018] [Indexed: 01/13/2023] Open
Abstract
Geographic atrophy (GA), the advanced form of dry age-related macular degeneration (AMD), is characterized by progressive loss of retinal pigment epithelium cells and photoreceptors in the setting of characteristic extracellular deposits and remains a serious unmet medical need. While genetic predisposition to AMD is dominated by polymorphisms in complement genes, it remains unclear how complement activation contributes to retinal atrophy. Here we demonstrate that complement is activated on photoreceptor outer segments (POS) in the retina peripheral to atrophic lesions associated with GA. When exposed to human serum following outer blood-retinal barrier breakdown, POS act as potent activators of the classical and alternative complement pathway. In mouse models of retinal degeneration, classical and alternative pathway complement activation on photoreceptors contributed to the loss of photoreceptor function. This was dependent on C5a-mediated recruitment of peripheral blood monocytes but independent of resident microglia. Genetic or pharmacologic inhibition of both classical and alternative complement C3 and C5 convertases was required to reduce progressive degeneration of photoreceptor rods and cones. Our study implicates systemic classical and alternative complement proteins and peripheral blood monocytes as critical effectors of localized retinal degeneration with potential relevance for the contribution of complement activation to GA.
Collapse
Affiliation(s)
- Kenneth J Katschke
- Department of Immunology, Genentech, Inc, South San Francisco, CA, 94080, USA
| | - Hongkang Xi
- Department of Immunology, Genentech, Inc, South San Francisco, CA, 94080, USA
| | - Christian Cox
- Department of Immunology, Genentech, Inc, South San Francisco, CA, 94080, USA
| | - Tom Truong
- Department of Translational Immunology, Genentech, Inc, South San Francisco, CA, 94080, USA
| | - Yann Malato
- Department of Translational Immunology, Genentech, Inc, South San Francisco, CA, 94080, USA
| | - Wyne P Lee
- Department of Translational Immunology, Genentech, Inc, South San Francisco, CA, 94080, USA
| | - Brent McKenzie
- Department of Translational Immunology, Genentech, Inc, South San Francisco, CA, 94080, USA
| | - Rommel Arceo
- Department of Pathology, Genentech, Inc, South San Francisco, CA, 94080, USA
| | - Jianhua Tao
- Department of Pathology, Genentech, Inc, South San Francisco, CA, 94080, USA
| | - Linda Rangell
- Department of Pathology, Genentech, Inc, South San Francisco, CA, 94080, USA
| | - Mike Reichelt
- Department of Pathology, Genentech, Inc, South San Francisco, CA, 94080, USA
| | - Lauri Diehl
- Department of Pathology, Genentech, Inc, South San Francisco, CA, 94080, USA
| | - Justin Elstrott
- Department of Biomedical Imaging, Genentech, Inc, South San Francisco, CA, 94080, USA
| | - Robby M Weimer
- Department of Biomedical Imaging, Genentech, Inc, South San Francisco, CA, 94080, USA
| | | |
Collapse
|
213
|
Pinna A, Zaccheddu F, Boscia F, Carru C, Solinas G. Homocysteine and risk of age-related macular degeneration: a systematic review and meta-analysis. Acta Ophthalmol 2018; 96:e269-e276. [PMID: 27966830 DOI: 10.1111/aos.13343] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 10/17/2016] [Indexed: 02/03/2023]
Abstract
There is still no agreement on total plasma homocysteine (tHcy) role in age-related macular degeneration (AMD), the leading cause of new blindness in industrialized countries. We performed a systematic review and meta-analysis of the published data on the correlation between tHcy and AMD. MEDLINE/PubMed and ISI Web of Sciences searches were performed according to MOOSE guidelines. Case-control studies were eligible for inclusion. Participants and controls were AMD patients and subjects without AMD. The main outcome measure was wet AMD. Homocysteine level was the main exposure variable. Data were pooled using a random-effects model. Twelve case-control studies were identified: 10 assessed wet AMD, four dry AMD, one early AMD, one late AMD, and one any AMD. As for wet AMD, there was a total of 453 cases and 514 controls. Mean tHcy was on average 1.1 μmol/l (95% confidence interval [CI] = 0.96-1.25) greater in wet AMD cases, but there was evidence of extreme between-study heterogeneity (p < 0.001, I2 = 91.8%). In a model homogenous for age, including six wet AMD studies (214 cases, 274 controls), mean tHcy was on average 0.58 μmol/l (95% CI = 0.35-0.73) greater in the case group, a not statistically significant result (p = 0.144) associated with moderate heterogeneity (I2 = 39.2%). Our meta-analysis indicates that there is some weak evidence that increased tHcy might be associated with wet AMD; however, this result should be interpreted cautiously, because of a marked between-study heterogeneity and the possible effect of publication bias. Future studies, preferably of cohort design, are necessary before any firm conclusions on the putative role of increased tHcy on AMD can be drawn.
Collapse
Affiliation(s)
- Antonio Pinna
- Department of Surgical, Microsurgical, and Medical Sciences; Ophthalmology Unit; University of Sassari; Sassari Italy
- Azienda Ospedaliero-Universitaria di Sassari; Sassari Italy
| | - Francesco Zaccheddu
- Department of Surgical, Microsurgical, and Medical Sciences; Ophthalmology Unit; University of Sassari; Sassari Italy
| | - Francesco Boscia
- Department of Surgical, Microsurgical, and Medical Sciences; Ophthalmology Unit; University of Sassari; Sassari Italy
- Azienda Ospedaliero-Universitaria di Sassari; Sassari Italy
| | - Ciriaco Carru
- Azienda Ospedaliero-Universitaria di Sassari; Sassari Italy
- Department of Biomedical Sciences; Section of Clinical Biochemistry; University of Sassari; Sassari Italy
| | - Giuliana Solinas
- Department of Biomedical Sciences; Laboratory of Epidemiology and Biostatistics; University of Sassari; Sassari Italy
| |
Collapse
|
214
|
Jiang J, Xu K, Wang L, Xin W, Zhao G, Huang M, Li S, Luan X, Fang J. Pharmacology study of a chimeric decoy receptor trap fusion protein on retina neovascularization by dual blockage of VEGF and FGF-2. Eur J Pharm Sci 2018; 121:251-259. [PMID: 29715501 DOI: 10.1016/j.ejps.2018.04.043] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 10/08/2017] [Accepted: 04/27/2018] [Indexed: 12/31/2022]
Abstract
BACKGROUND Clinical anti-vascular epithelial growth factor (VEGF) therapy trials faced a major challenge due to upregulated expression of other pro-angiogenic factors, such as fibroblast growth factor-2 (FGF-2). RC28, a novel recombinant dual decoy receptor IgG1 Fc-fusion protein, can block VEGFA and FGF-2 simultaneously. It is designed for the treatment of neovascular age-related macular degeneration and other pathological ocular neovascularization. The present study investigated the prevention efficacy of RC28 on choroidal neovascularization (CNV) in a monkey model and compared to the other mono VEGF antagonists; biodistribution and pharmacokinetics performance were also investigated. METHODS ELISA and endothelial cell proliferation, migration, and tubule formation assay evaluated the bioactivity of RC28 in vitro, and an initial comparison was made among the mono target antagonists, Bevacizumab (Avastin), Ranibizumab (Lucentis), Aflibercept (EYLEA), Conbercept (KH902), and Ranibizumab (Lucentis). Laser-induced CNV in monkeys, and both VEGF and FGF-2 serum levels were detected in animals before and after the CNV model were induced. RC28 prevention efficacy was compared to other VEGF antagonists on CNV with respect to the incidence of CNV and several ophthalmic examinations. Ocular and systemic levels of RC28 were analyzed by 89Zr-labeled RC28 after single intravitreal administration for the biodistribution and pharmacokinetic profiles. RESULTS RC28 is a unique fusion protein with high affinity to both VEGF and FGF-2, and beneficial to in vitro and in vivo bioactivity. The in vivo pharmacological studies demonstrated that the incidence of CNV formation was largely reduced in RC28 treatment groups with a low dosage as compared to other VEGF antagonist control groups. Furthermore, traces of RC28 were detected as dispersing from eyeballs to the liver after 20 days, and a prolonged half-time pharmacokinetic profile was exhibited.
Collapse
Affiliation(s)
- Jing Jiang
- Department of Pharmacology, Binzhou Medical University, Yantai 256603, Shandong, China.
| | - Ke Xu
- Department of Pharmacology and Toxicology, Beijing, Institute of Radiation Medicine, Beijing 100850, China
| | - Ling Wang
- RemeGen, Ltd, Yantai 264000, Shandong, China
| | - Wei Xin
- RemeGen, Ltd, Yantai 264000, Shandong, China
| | - Guorui Zhao
- RemeGen, Ltd, Yantai 264000, Shandong, China
| | - Min Huang
- RemeGen, Ltd, Yantai 264000, Shandong, China
| | - Shenjun Li
- RemeGen, Ltd, Yantai 264000, Shandong, China
| | | | - Jianmin Fang
- School of Life Science and Technology, Tongji University, Shanghai 200092, China
| |
Collapse
|
215
|
Zhou M, Luo J, Zhang H. Role of Sirtuin 1 in the pathogenesis of ocular disease (Review). Int J Mol Med 2018; 42:13-20. [PMID: 29693113 DOI: 10.3892/ijmm.2018.3623] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 03/29/2018] [Indexed: 11/06/2022] Open
Abstract
Sirtuin (SIRT)1, a member of the SIRT family, is a highly conserved NAD+‑dependent histone deacetylase, which has a regulatory role in numerous physiological and pathological processes by removing acetyl groups from various proteins. SIRT1 controls the activity of numerous transcription factors and cofactors, which impacts the downstream gene expression, and eventually alleviates oxidative stress and associated damage. Numerous studies have revealed that dysfunction of SIRT1 is linked with ocular diseases, including cataract, age‑associated macular degeneration, diabetic retinopathy and glaucoma, while ectopic upregulation of SIRT1 protects against various ocular diseases. In the present review, the significant role of SIRT1 and the potential therapeutic value of modulating SIRT1 expression in ocular development and eye diseases is summarized.
Collapse
Affiliation(s)
- Mengwen Zhou
- Department of Ophthalmology, Hunan Clinical Research Center of Ophthalmic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Jing Luo
- Department of Ophthalmology, Hunan Clinical Research Center of Ophthalmic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Huiming Zhang
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| |
Collapse
|
216
|
Salas M, Augustin M, Felberer F, Wartak A, Laslandes M, Ginner L, Niederleithner M, Ensher J, Minneman MP, Leitgeb RA, Drexler W, Levecq X, Schmidt-Erfurth U, Pircher M. Compact akinetic swept source optical coherence tomography angiography at 1060 nm supporting a wide field of view and adaptive optics imaging modes of the posterior eye. BIOMEDICAL OPTICS EXPRESS 2018; 9:1871-1892. [PMID: 29675326 PMCID: PMC5905931 DOI: 10.1364/boe.9.001871] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 03/13/2018] [Accepted: 03/13/2018] [Indexed: 05/25/2023]
Abstract
Imaging of the human retina with high resolution is an essential step towards improved diagnosis and treatment control. In this paper, we introduce a compact, clinically user-friendly instrument based on swept source optical coherence tomography (SS-OCT). A key feature of the system is the realization of two different operation modes. The first operation mode is similar to conventional OCT imaging and provides large field of view (FoV) images (up to 45° × 30°) of the human retina and choroid with standard resolution. The second operation mode enables it to optically zoom into regions of interest with high transverse resolution using adaptive optics (AO). The FoV of this second operation mode (AO-OCT mode) is 3.0° × 2.8° and enables the visualization of individual retinal cells such as cone photoreceptors or choriocapillaris. The OCT engine is based on an akinetic swept source at 1060 nm and provides an A-scan rate of 200 kHz. Structural as well as angiographic information can be retrieved from the retina and choroid in both operational modes. The capabilities of the prototype are demonstrated in healthy and diseased eyes.
Collapse
Affiliation(s)
- Matthias Salas
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
- Christian Doppler Laboratory for Innovative Optical Imaging and Its Translation to Medicine, Medical University of Vienna, Vienna, Austria
| | - Marco Augustin
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
| | | | - Andreas Wartak
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
| | - Marie Laslandes
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
| | - Laurin Ginner
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
- Christian Doppler Laboratory for Innovative Optical Imaging and Its Translation to Medicine, Medical University of Vienna, Vienna, Austria
| | - Michael Niederleithner
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
| | - Jason Ensher
- INSIGHT Photonic Solutions, Inc., Lafayette, CO, USA
| | | | - Rainer A. Leitgeb
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
- Christian Doppler Laboratory for Innovative Optical Imaging and Its Translation to Medicine, Medical University of Vienna, Vienna, Austria
| | - Wolfgang Drexler
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
| | | | - Ursula Schmidt-Erfurth
- Department of Ophthalmology and Optometry, Medical University of Vienna, Vienna, Austria
| | - Michael Pircher
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
217
|
Tang CZ, Li KR, Yu Q, Jiang Q, Yao J, Cao C. Activation of Nrf2 by Ginsenoside Rh3 protects retinal pigment epithelium cells and retinal ganglion cells from UV. Free Radic Biol Med 2018; 117:238-246. [PMID: 29427790 DOI: 10.1016/j.freeradbiomed.2018.02.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 01/11/2018] [Accepted: 02/02/2018] [Indexed: 12/20/2022]
Abstract
Excessive Ultra-violet (UV) radiation shall induce damages to resident retinal pigment epithelium (RPE) cells (RPEs) and retinal ganglion cells (RGCs). Here we tested the potential activity of Ginsenoside Rh3 ("Rh3") against the process. In cultured human RPEs and RGCs, pretreatment with Rh3 inhibited UV-induced reactive oxygen species (ROS) production and following apoptotic/non-apoptotic cell death. Rh3 treatment in retinal cells induced nuclear-factor-E2-related factor 2 (Nrf2) activation, which was evidenced by Nrf2 protein stabilization and its nuclear translocation, along with transcription of antioxidant responsive element (ARE)-dependent genes (HO1, NOQ1 and GCLC). Nrf2 knockdown by targeted-shRNA almost abolished Rh3-induced retinal cell protection against UV. Further studies found that Rh3 induced microRNA-141 ("miR-141") expression, causing downregulation of its targeted gene Keap1 in RPEs and RGCs. On the other hand, Rh3-induced Nrf2 activation and retinal cell protection were largely attenuated by the miR-141's inhibitor, antagomiR-141. In vivo, intravitreal injection of Rh3 inhibited retinal dysfunction by light damage in mice. Rh3 intravitreal injection also induced miR-141 expression, Keap1 downregulation and Nrf2 activation in mouse retinas. We conclude that Rh3 protects retinal cells from UV via activating Nrf2 signaling.
Collapse
Affiliation(s)
- Chun-Zhou Tang
- The Affiliated Eye Hospital of Nanjing Medical University, Nanjing, China; Ophthalmology Department, Jiangsu Jiangyin people's Hospital, Jiangyin, China
| | - Ke-Ran Li
- The Affiliated Eye Hospital of Nanjing Medical University, Nanjing, China
| | - Qing Yu
- The Affiliated Eye Hospital of Nanjing Medical University, Nanjing, China
| | - Qin Jiang
- The Affiliated Eye Hospital of Nanjing Medical University, Nanjing, China.
| | - Jin Yao
- The Affiliated Eye Hospital of Nanjing Medical University, Nanjing, China.
| | - Cong Cao
- The Affiliated Eye Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China; Center of Translational Medicine, The First People Hospital of Zhangjiagang City, Soochow University, Suzhou, China; North District, The Municipal Hospital of Suzhou, Suzhou, China.
| |
Collapse
|
218
|
Dieguez HH, Romeo HE, González Fleitas MF, Aranda ML, Milne GA, Rosenstein RE, Dorfman D. Superior cervical gangliectomy induces non-exudative age-related macular degeneration in mice. Dis Model Mech 2018; 11:dmm.031641. [PMID: 29361515 PMCID: PMC5894943 DOI: 10.1242/dmm.031641] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 11/28/2017] [Indexed: 12/20/2022] Open
Abstract
Non-exudative age-related macular degeneration, a prevalent cause of blindness, is a progressive and degenerative disease characterized by alterations in Bruch's membrane, retinal pigment epithelium, and photoreceptors exclusively localized in the macula. Although experimental murine models exist, the vast majority take a long time to develop retinal alterations and, in general, these alterations are ubiquitous, with many resulting from non-eye-specific genetic manipulations; additionally, most do not always reproduce the hallmarks of human age-related macular degeneration. Choroid vessels receive sympathetic innervation from the superior cervical ganglion, which, together with the parasympathetic system, regulates blood flow into the choroid. Choroid blood flow changes have been involved in age-related macular degeneration development and progression. At present, no experimental models take this factor into account. The aim of this work was to analyze the effect of superior cervical gangliectomy (also known as ganglionectomy) on the choroid, Bruch's membrane, retinal pigment epithelium and retina. Adult male C57BL/6J mice underwent unilateral superior cervical gangliectomy and a contralateral sham procedure. Although superior cervical gangliectomy induced ubiquitous choroid and choriocapillaris changes, it induced Bruch's membrane thickening, loss of retinal pigment epithelium melanin content and retinoid isomerohydrolase, the appearance of drusen-like deposits, and retinal pigment epithelium and photoreceptor atrophy, exclusively localized in the temporal side. Moreover, superior cervical gangliectomy provoked a localized increase in retinal pigment epithelium and photoreceptor apoptosis, and a decline in photoreceptor electroretinographic function. Therefore, superior cervical gangliectomy recapitulated the main features of human non-exudative age-related macular degeneration, and could become a new experimental model of dry age-related macular degeneration, and a useful platform for developing new therapies. Summary: Ubiquitous alteration of choroid circulation causes localized retinal alterations in mice that are similar to human non-exudative age-related macular degeneration, thus providing a new potential experimental model of the disease.
Collapse
Affiliation(s)
- Hernán H Dieguez
- Laboratory of Retinal Neurochemistry and Experimental Ophthalmology, Department of Human Biochemistry, School of Medicine/CEFyBO, University of Buenos Aires/CONICET, Buenos Aires C1121ABG, Argentina
| | - Horacio E Romeo
- Faculty of Medical Sciences, Pontifical Catholic University of Argentina, BIOMED/UCA/CONICET, Buenos Aires C1107AFB, Argentina
| | - María F González Fleitas
- Laboratory of Retinal Neurochemistry and Experimental Ophthalmology, Department of Human Biochemistry, School of Medicine/CEFyBO, University of Buenos Aires/CONICET, Buenos Aires C1121ABG, Argentina
| | - Marcos L Aranda
- Laboratory of Retinal Neurochemistry and Experimental Ophthalmology, Department of Human Biochemistry, School of Medicine/CEFyBO, University of Buenos Aires/CONICET, Buenos Aires C1121ABG, Argentina
| | - Georgia A Milne
- Laboratory of Retinal Neurochemistry and Experimental Ophthalmology, Department of Human Biochemistry, School of Medicine/CEFyBO, University of Buenos Aires/CONICET, Buenos Aires C1121ABG, Argentina
| | - Ruth E Rosenstein
- Laboratory of Retinal Neurochemistry and Experimental Ophthalmology, Department of Human Biochemistry, School of Medicine/CEFyBO, University of Buenos Aires/CONICET, Buenos Aires C1121ABG, Argentina
| | - Damián Dorfman
- Laboratory of Retinal Neurochemistry and Experimental Ophthalmology, Department of Human Biochemistry, School of Medicine/CEFyBO, University of Buenos Aires/CONICET, Buenos Aires C1121ABG, Argentina
| |
Collapse
|
219
|
Gong YQ, Huang W, Li KR, Liu YY, Cao GF, Cao C, Jiang Q. SC79 protects retinal pigment epithelium cells from UV radiation via activating Akt-Nrf2 signaling. Oncotarget 2018; 7:60123-60132. [PMID: 27517753 PMCID: PMC5312373 DOI: 10.18632/oncotarget.11164] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 07/20/2016] [Indexed: 12/11/2022] Open
Abstract
Excessive Ultra-violet (UV) radiation causes oxidative damages and apoptosis in retinal pigment epithelium (RPE) cells. Here we tested the potential activity of SC79, a novel small molecule activator of Akt, against the process. We showed that SC79 activated Akt in primary and established (ARPE-19 line) RPE cells. It protected RPE cells from UV damages possibly via inhibiting cell apoptosis. Akt inhibition, via an Akt specific inhibitor (MK-2206) or Akt1 shRNA silence, almost abolished SC79-induced RPE cytoprotection. Further studies showed that SC79 activated Akt-dependent NF-E2-related factor 2 (Nrf2) signaling and inhibited UV-induced oxidative stress in RPE cells. Reversely, Nrf2 shRNA knockdown or S40T mutation attenuated SC79-induced anti-UV activity. For the in vivo studies, we showed that intravitreal injection of SC79 significantly protected mouse retina from light damages. Based on these results, we suggest that SC79 protects RPE cells from UV damages possibly via activating Akt-Nrf2 signaling axis.
Collapse
Affiliation(s)
- Yi-Qing Gong
- The Affiliated Eye Hospital of Nanjing Medical University, Nanjing, China.,Ophthalmology Department, Zhenjiang First People's Hospital, Zhenjiang, China
| | - Wei Huang
- The Affiliated Eye Hospital of Nanjing Medical University, Nanjing, China
| | - Ke-Ran Li
- The Affiliated Eye Hospital of Nanjing Medical University, Nanjing, China
| | - Yuan-Yuan Liu
- Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, China
| | - Guo-Fan Cao
- The Affiliated Eye Hospital of Nanjing Medical University, Nanjing, China
| | - Cong Cao
- Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, China
| | - Qin Jiang
- The Affiliated Eye Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
220
|
Chen Y, Zhong M, Yuan G, Peng H. Interleukin-17 induces angiogenesis in vitro via CXCL8 and CCL2 in retinal pigment epithelium. Mol Med Rep 2018; 17:4627-4632. [PMID: 29363728 DOI: 10.3892/mmr.2018.8460] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 11/30/2017] [Indexed: 11/06/2022] Open
Abstract
Interleukin-17 (IL-17) is a major pro-inflammatory cytokine involved in choroidal endothelial cell (CEC) angiogenesis. Proteins expressed by the retinal pigment epithelium (RPE) may contribute to CEC angiogenesis. The ability of IL‑17 to promote proliferation, migration and capillary‑like structure formation in CECs was investigated by stimulating the RPE in vitro. CECs were cultured in a conditioned medium (CM) with IL‑17 (IL‑17‑CM) or without IL‑17 (CM) obtained from the supernatant of an ARPE‑19 cell line. The pro‑angiogenic role of IL‑17‑CM on CECs was investigated with water‑soluble tetrazolium 1 analysis, wound healing and Matrigel matrix tube formation assays. The expression level of vascular endothelial growth factor was detected by enzyme‑linked immunosorbent assay in RPE cells treated with or without IL‑17. Ras‑related C3botulinum toxin substrate 1 (Rac1) and Ras homolog gene family member A (RhoA) activities were analyzed by pull‑down assays. IL‑17‑CM significantly enhanced tube formation and increased the migration distance in CECs in comparison with CM. This effect was diminished by neutralizing C‑C motif chemokine 2 (CCL2) and C‑X‑C motif chemokine ligand 8 (CXCL8) expression in IL‑17‑CM, with a concomitant downregulation of Rac1 and RhoA activity in CECs. In conclusion, it was demonstrated that IL‑17 mediated the expression of CCL2 and CXCL8 in RPE cells, resulting in increased migration and tube formation in human CECs.
Collapse
Affiliation(s)
- Ying Chen
- Department of Ophthalmology, First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, Chongqing 400016, P.R. China
| | - Murui Zhong
- Department of Ophthalmology, 452 Hospital of People's Liberation Army, Chengdu, Sichuan 610065, P.R. China
| | - Gangxiang Yuan
- Department of Ophthalmology, First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, Chongqing 400016, P.R. China
| | - Hui Peng
- Department of Ophthalmology, First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, Chongqing 400016, P.R. China
| |
Collapse
|
221
|
Hu H, Hao L, Tang C, Zhu Y, Jiang Q, Yao J. Activation of KGFR-Akt-mTOR-Nrf2 signaling protects human retinal pigment epithelium cells from Ultra-violet. Biochem Biophys Res Commun 2018; 495:2171-2177. [DOI: 10.1016/j.bbrc.2017.12.078] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 12/14/2017] [Indexed: 11/24/2022]
|
222
|
Koh LHL, Agrawal R, Khandelwal N, Sai Charan L, Chhablani J. Choroidal vascular changes in age-related macular degeneration. Acta Ophthalmol 2017; 95:e597-e601. [PMID: 28391615 DOI: 10.1111/aos.13399] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 12/19/2016] [Indexed: 01/21/2023]
Abstract
PURPOSE To assess the choroidal vascular changes using choroidal vascularity index (CVI) in patients with age-related macular degeneration (AMD) compared to controls. METHODS Enhanced depth imaging (EDI) optical coherence tomography (OCT) scans of 64 patients with unilateral or bilateral AMD were obtained. Images with a poorly demarcated choroidal-scleral interface (CSI) were excluded from the analysis. Foveal scans of 63 AMD eyes and 35 'normal fellow' eyes were analysed. Images of 30 eyes from 18 age-matched healthy subjects were included as controls. Choroidal vascularity index (CVI) was derived from binarization of EDI OCT images, using fiji software. RESULTS The mean age was 56.50 ± 5.50 years for AMD patients and 52.25 ± 6.75 years for controls. All patients were treatment naïve. Subfoveal choroidal thickness (SFCT) in AMD, 'normal fellow' eyes and controls was 314.02 ± 78.80 μm, 300.88 ± 53.85 μm and 278.5 ± 65.31 μm, respectively. Choroidal vascularity index (CVI) in AMD, 'normal fellow' eyes and controls was 64.04 ± 2.43%, 64.66 ± 2.25% and 66.07 ± 1.72%, respectively. Choroidal vascularity index (CVI) of both AMD and 'normal fellow' eyes was significantly lower compared to controls (p < 0.0001 and p = 0.007). The SFCT of AMD eye was not found to be significantly different from 'normal fellow eyes' (p = 0.45). CONCLUSION There was no statistical difference in SFCT, but CVI was significantly lower in patients with AMD. Choroidal vascularity index (CVI) was also lower in 'normal fellow' AMD eyes as compared to controls. This suggests possible reduction in choroidal vascularity in eyes with AMD and also to a certain extent in the 'normal fellow' eyes without phenotypical manifestations and may suggest underlying choroidal morphological change leading to wet AMD.
Collapse
Affiliation(s)
- Lilian Hui Li Koh
- National Healthcare Group Eye Institute; Tan Tock Seng Hospital; Singapore Singapore
| | - Rupesh Agrawal
- National Healthcare Group Eye Institute; Tan Tock Seng Hospital; Singapore Singapore
| | - Neha Khandelwal
- National Healthcare Group Eye Institute; Tan Tock Seng Hospital; Singapore Singapore
| | - Labishetty Sai Charan
- National Healthcare Group Eye Institute; Tan Tock Seng Hospital; Singapore Singapore
| | | |
Collapse
|
223
|
Kandasamy R, Wickremasinghe S, Guymer R. New Treatment Modalities for Geographic Atrophy. Asia Pac J Ophthalmol (Phila) 2017; 6:508-513. [PMID: 28905539 DOI: 10.22608/apo.2017262] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Age‑related macular degeneration (AMD) is a significant cause of global visual morbidity and is projected to affect 288 million people by the year 2040. The advent of treatment with anti‒vascular endothelial growth factor (anti‑VEGF) drugs has revolutionized the treatment of neovascular AMD (nAMD) but there have been no similar breakthroughs for the treatment of geographic atrophy (GA) to retard its progression. The advancements in imaging and new understanding of disease mechanisms, based on molecular and genetic models, have paved the way for the development of novel experimental treatment options for GA that aim to cater to a thus far largely unmet need. This review paper focuses on the recent clinical trials of new treatment options for slowing GA progression rates with emphasis on the agents that are currently undergoing, or have already undergone, significant clinical trial testing. Several new groups of drugs, including those targeting the complement cascade and agents considered as neuroprotective, have shown some promising results and could potentially pave the way forward in the treatment of this devastating disease.
Collapse
Affiliation(s)
- Rathika Kandasamy
- Centre for Eye Research Australia, Department of Surgery (Ophthalmology), University of Melbourne, Royal Victorian Eye and Ear, Hospital, Melbourne, Australia
| | - Sanjeewa Wickremasinghe
- Centre for Eye Research Australia, Department of Surgery (Ophthalmology), University of Melbourne, Royal Victorian Eye and Ear, Hospital, Melbourne, Australia
| | - Robyn Guymer
- Centre for Eye Research Australia, Department of Surgery (Ophthalmology), University of Melbourne, Royal Victorian Eye and Ear, Hospital, Melbourne, Australia
| |
Collapse
|
224
|
MIND4-17 protects retinal pigment epithelium cells and retinal ganglion cells from UV. Oncotarget 2017; 8:89793-89801. [PMID: 29163788 PMCID: PMC5685709 DOI: 10.18632/oncotarget.21131] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 09/03/2017] [Indexed: 01/01/2023] Open
Abstract
Nrf2 activation would efficiently protect retinal cells from UV radiation (UVR). Recent studies have developed a Nrf2-targeting thiazole-containing compound MIND4-17, which activates Nrf2 through blocking its association with Keap1. In the current study, we demonstrated that pretreatment with MIND4-17 efficiently protected retinal pigment epithelium (RPE) cells (RPEs) and retinal ganglion cells (RGCs) from UVR. UVR-induced apoptosis in the retinal cells was also largely attenuated by MIND4-17 pretreatment. MIND4-17 presumably separated Nrf2 from Keap1, allowing its stabilization and accumulation in retinal cells, which then translocated to cell nuclei and promoted transcription of ARE-dependent anti-oxidant genes, including HO1, NQO1 and GCLM. Significantly, shRNA-mediated knockdown of Nrf2 almost completely abolished MIND4-17-induced cytoprotection against UVR. Further studies showed that MIND4-17 largely ameliorated UVR-induced ROS production, lipid peroxidation and DNA damages in RPEs and RGCs. Together, MIND4-17 protects retinal cells from UVR by activating Nrf2 signaling.
Collapse
|
225
|
Hu Y, Yang B, Xu Y, Jiang L, Tsui CK, Liang X. FK506 suppresses hypoxia‑induced inflammation and protects tight junction function via the CaN‑NFATc1 signaling pathway in retinal microvascular epithelial cells. Mol Med Rep 2017; 16:6974-6980. [PMID: 28901449 DOI: 10.3892/mmr.2017.7475] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 05/24/2017] [Indexed: 11/06/2022] Open
Abstract
The present study aimed to identify whether FK506 suppresses hypoxia‑induced inflammation and protects tight junction function via the calcineurin‑nuclear factor of activated T‑cells 1 (CaN‑NFATc1) signaling pathway in mouse retinal microvascular endothelial cells (mRMECs). The mRMECs were treated with FK506 at different concentrations following the induction of hypoxia. Trans‑epithelial electrical resistance (TEER) and cell permeability were examined to measure the integrity of the tight junctions. The concentrations of inflammatory cytokines were measured using reverse transcription‑quantitative polymerase chain reaction analysis and enzyme‑linked immunosorbent assays. The protein expression levels of zonula occludens‑1 (ZO‑1) and nuclear factor of activated T‑cell 1 (NFATc1) were identified using immunofluorescent microscopy and western blot analysis. The TEER value was decreased following hypoxia, but increased following treatment with FK506 (1 and 10 µM) for 24 and 48 h. The protein expression of ZO‑1 was also increased following FK506 treatment for 24 h at 1 and 10 µM. By contrast, following treatment with FK506 (1 and 10 µM) for 24 and 48 h, the elevated cell permeability in the hypoxia group was significantly downregulated. Similarly, the concentrations of inflammatory cytokines, including cyclooxygenase‑2, inducible nitric oxide synthase, monocyte chemoattractant protein‑1, interleukin‑6, intercellular adhesion molecule‑1 and vascular cell adhesion molecule‑1, were downregulated following treatment with FK506 for 24 h at 1 and 10 µM. Following treatment with FK506, the level of total NFATc1 was downregulated and the level of phosphorylated NFATc1 was upregulated. Taken together, FK506 suppressed injury to the tight junctions and downregulated the expression of inflammatory cytokines in hypoxia‑induced mRMECs via the CaN‑NFATc1 signaling pathway. This suggests a potentially effective therapy for hypoxia‑induced retinal microangiopathy.
Collapse
Affiliation(s)
- Yaguang Hu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat‑sen University, Guangzhou, Guangdong 510000, P.R. China
| | - Boyu Yang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat‑sen University, Guangzhou, Guangdong 510000, P.R. China
| | - Yue Xu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat‑sen University, Guangzhou, Guangdong 510000, P.R. China
| | - Li Jiang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat‑sen University, Guangzhou, Guangdong 510000, P.R. China
| | - Ching-Kit Tsui
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat‑sen University, Guangzhou, Guangdong 510000, P.R. China
| | - Xiaoling Liang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat‑sen University, Guangzhou, Guangdong 510000, P.R. China
| |
Collapse
|
226
|
Cheng Q, Saaddine JB, Klein R, Rothenberg R, Chou CF, Il'yasova D. Early Age-related Macular Degeneration with Cardiovascular and Renal Comorbidities: An Analysis of the National Health and Nutrition Examination Survey, 2005-2008. Ophthalmic Epidemiol 2017; 24:413-419. [PMID: 28891729 DOI: 10.1080/09286586.2017.1337911] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
PURPOSE A cross sectional study was designed to examine the relationship of early age-related macular degeneration (AMD) with comorbidities of cardiovascular and renal conditions in the representative population using National Health and Nutrition Examination Survey (NHANES), 2005-2008. METHODS Participants (≥40 years) who underwent retinal photography were included. Early AMD was defined by the retinal digital images. The comorbidities were self-reported stroke and heart disease (HD), including angina pectoris (AP), coronary heart disease (CHD), congestive heart failure (CHF), and myocardial infarction (MI). Chronic kidney disease (CKD) was determined based on self-report, estimation of glomerular filtration rate (GFR), or the level of urine albumin. RESULTS The age-adjusted odds ratio (OR) and 95% CI for having early AMD for persons with the selected conditions were: 2.6 (1.9, 3.6) for any type of HD. When the conditions were considered separately, ORs (95% CIs) were: 2.0 (1.2, 3.4) for AP; 2.5 (1.6, 3.8) for CHD; 2.4 (1.6, 3.6) for MI; 2.3 (1.3, 3.9) for CHF; 3.3 (2.2, 5.0) for stroke; and 2.4 (1.8, 3.2) for CKD. Covariable-adjusted ORs (AOR) were attenuated for all examined conditions, but remained statistically significant. Having any single condition (AOR [95%CI]: 2.7 [1.5, 4.8]) was significantly associated with early AMD, as was having ≥ 2 conditions (AOR [95%CI]: 5.2 [3.0, 9.0]). The strongest association was between early AMD and the combination of HD and stroke (AOR [95% CI]: 6.3 [2.9, 13.8]). CONCLUSION Cardiovascular and renal comorbidities are associated with early AMD in a representative sample of the US general population.
Collapse
Affiliation(s)
- Qi Cheng
- a Department of Epidemiology and Biostatistics , Georgia State University School of Public Health , Atlanta , Georgia , USA
| | - Jinan B Saaddine
- b Division of Diabetes Translation , Centers for Disease Control and Prevention , Atlanta , Georgia , USA
| | - Ronald Klein
- c Department of Ophthalmology and Visual Sciences , University of Wisconsin School of Medicine and Public Health , Madison , Wisconsin , USA
| | - Richard Rothenberg
- a Department of Epidemiology and Biostatistics , Georgia State University School of Public Health , Atlanta , Georgia , USA
| | - Chiu-Fang Chou
- b Division of Diabetes Translation , Centers for Disease Control and Prevention , Atlanta , Georgia , USA
| | - Dora Il'yasova
- a Department of Epidemiology and Biostatistics , Georgia State University School of Public Health , Atlanta , Georgia , USA
| |
Collapse
|
227
|
Hu Y, Lu X, Xu Y, Lu L, Yu S, Cheng Q, Yang B, Tsui CK, Ye D, Huang J, Liang X. Salubrinal attenuated retinal neovascularization by inhibiting CHOP-HIF1α-VEGF pathways. Oncotarget 2017; 8:77219-77232. [PMID: 29100382 PMCID: PMC5652775 DOI: 10.18632/oncotarget.20431] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 06/12/2017] [Indexed: 12/30/2022] Open
Abstract
Retinal neovascularization (RNV) related disease is the leading cause of irreversible blindness in the world. The aim of this study is to identify whether salubrinal could attenuate RNV by inhibiting CCAAT/enhancer-binding protein (C/EBP) homologous protein (CHOP)- hypoxia inducible factors 1α (HIF1α) -vascular endothelial growth factor (VEGF) pathways in both mouse retinal microvascular endothelial cells (mRMECs) and oxygen-induced retinopathy (OIR) mouse model. After being treated with salubrinal (20μmol/L) or CHOP-siRNA, mRMECs were exposed to a hypoxia environment. OIR mice were intraperitoneally injected with salubrinal (0.5 mg/kg/day) from P12 to P17. With salubrinal or CHOP-siRNA treatment, the elevated CHOP protein and mRNA levels in hypoxia-induced mRMECs were significantly decreased. HIF1α-VEGF pathways were activated under hypoxia condition, then HIF1α protein was degraded and VEGF secretion was down-regulated after salubrinal or CHOP-siRNA treatment. In OIR mice, the areas of RNV were markedly decreased with salubrinal treatment. Moreover, elevated expressions of CHOP, HIF1α and VEGF in retinas of OIR mice were all reduced after salubrinal treatment. It suggested that salubrinal attenuated RNV in mRMECs and OIR mice by inhibiting CHOP-HIF1α-VEGF pathways and could be a potential therapeutic target for hypoxia-induced retinal microangiopathy.
Collapse
Affiliation(s)
- Yaguang Hu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510000, Guangdong Province, China
| | - Xi Lu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510000, Guangdong Province, China
| | - Yue Xu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510000, Guangdong Province, China
| | - Lin Lu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510000, Guangdong Province, China
| | - Shanshan Yu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510000, Guangdong Province, China
| | - Qiaochu Cheng
- Department of Ophthalmology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, China
| | - Boyu Yang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510000, Guangdong Province, China
| | - Ching-Kit Tsui
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510000, Guangdong Province, China
| | - Dan Ye
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510000, Guangdong Province, China
| | - Jingjing Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510000, Guangdong Province, China
| | - Xiaoling Liang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510000, Guangdong Province, China
| |
Collapse
|
228
|
Schlecht A, Leimbeck SV, Jägle H, Feuchtinger A, Tamm ER, Braunger BM. Deletion of Endothelial Transforming Growth Factor-β Signaling Leads to Choroidal Neovascularization. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:2570-2589. [PMID: 28823871 DOI: 10.1016/j.ajpath.2017.06.018] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 06/12/2017] [Accepted: 06/29/2017] [Indexed: 12/13/2022]
Abstract
The molecular pathogenesis of choroidal neovascularization (CNV), an angiogenic process that critically contributes to vision loss in age-related macular degeneration, is unclear. Herein, we analyzed the role of transforming growth factor (TGF)-β signaling for CNV formation by generating a series of mutant mouse models with induced conditional deletion of TGF-β signaling in the entire eye, the retinal pigment epithelium (RPE), or the vascular endothelium. Deletion of TGF-β signaling in the eye caused CNV, irrespectively if it was ablated in newborn or 3-week-old mice. Areas of CNV showed photoreceptor degeneration, multilayered RPE, basal lamina deposits, and accumulations of monocytes/macrophages. The changes progressed, leading to marked structural and functional alterations of the retina. Although the specific deletion of TGF-β signaling in the RPE caused no obvious changes, specific deletion in vascular endothelial cells caused CNV and a phenotype similar to that observed after the deletion in the entire eye. We conclude that impairment of TGF-β signaling in the vascular endothelium of the eye is sufficient to trigger CNV formation. Our findings highlight the importance of TGF-β signaling as a key player in the development of ocular neovascularization and indicate a fundamental role of TGF-β signaling in the pathogenesis of age-related macular degeneration.
Collapse
Affiliation(s)
- Anja Schlecht
- Institute of Human Anatomy and Embryology, University of Regensburg, Regensburg, Germany
| | - Sarah V Leimbeck
- Institute of Human Anatomy and Embryology, University of Regensburg, Regensburg, Germany
| | - Herbert Jägle
- Department of Ophthalmology, University Clinic Regensburg, Regensburg, Germany
| | - Annette Feuchtinger
- Research Unit Analytical Pathology, Helmholtz Zentrum Munich, Munich, Germany
| | - Ernst R Tamm
- Institute of Human Anatomy and Embryology, University of Regensburg, Regensburg, Germany.
| | - Barbara M Braunger
- Institute of Human Anatomy and Embryology, University of Regensburg, Regensburg, Germany.
| |
Collapse
|
229
|
miRNA-141 attenuates UV-induced oxidative stress via activating Keap1-Nrf2 signaling in human retinal pigment epithelium cells and retinal ganglion cells. Oncotarget 2017; 8:13186-13194. [PMID: 28061435 PMCID: PMC5355087 DOI: 10.18632/oncotarget.14489] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 12/07/2016] [Indexed: 12/30/2022] Open
Abstract
Activation of NF-E2-related factor 2 (Nrf2) signaling could protect cells from ultra violet (UV) radiation. We aim to provoke Nrf2 activation via downregulating its inhibitor Keap1 by microRNA-141 (“miR-141”). In both human retinal pigment epithelium cells (RPEs) and retinal ganglion cells (RGCs), forced-expression of miR-141 downregulated Keap1, causing Nrf2 stabilization, accumulation and nuclear translocation, which led to transcription of multiple antioxidant-responsive element (ARE) genes (HO1, NOQ1 and GCLC). Further, UV-induced reactive oxygen species (ROS) production and cell death were significantly attenuated in miR-141-expressing RPEs and RGCs. On the other hand, depletion of miR-141 via expressing its inhibitor antagomiR-141 led to Keap1 upregulation and Nrf2 degradation, which aggravated UV-induced death of RPEs and RGCs. Significantly, Nrf2 shRNA knockdown almost abolished miR-141-mediated cytoprotection against UV in RPEs. These results demonstrate that miR-141 targets Keap1 to activate Nrf2 signaling, which protects RPEs and RGCs from UV radiation.
Collapse
|
230
|
Moore NA, Bracha P, Hussain RM, Morral N, Ciulla TA. Gene therapy for age-related macular degeneration. Expert Opin Biol Ther 2017; 17:1235-1244. [PMID: 28726562 DOI: 10.1080/14712598.2017.1356817] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION In neovascular age related macular degeneration (nAMD), gene therapy to chronically express anti-vascular endothelial growth factor (VEGF) proteins could ameliorate the treatment burden of chronic intravitreal therapy and improve limited visual outcomes associated with 'real world' undertreatment. Areas covered: In this review, the authors assess the evolution of gene therapy for AMD. Adeno-associated virus (AAV) vectors can transduce retinal pigment epithelium; one such early application was a phase I trial of AAV2-delivered pigment epithelium derived factor gene in advanced nAMD. Subsequently, gene therapy for AMD shifted to the investigation of soluble fms-like tyrosine kinase-1 (sFLT-1), an endogenously expressed VEGF inhibitor, binding and neutralizing VEGF-A. After some disappointing results, research has centered on novel vectors, including optimized AAV2, AAV8 and lentivirus, as well as genes encoding other anti-angiogenic proteins, including ranibizumab, aflibercept, angiostatin and endostatin. Also, gene therapy targeting the complement system is being investigated for geographic atrophy due to non-neovascular AMD. Expert opinion: The success of gene therapy for AMD will depend on the selection of the most appropriate therapeutic protein and its level of chronic expression. Future investigations will center on optimizing vector, promoter and delivery methods, and evaluating the risks of the chronic expression of anti-angiogenic or anti-complement proteins.
Collapse
Affiliation(s)
- Nicholas A Moore
- a Department of Ophthalmology , Indiana University School of Medicine , Indianapolis , IN , USA
| | - Peter Bracha
- a Department of Ophthalmology , Indiana University School of Medicine , Indianapolis , IN , USA
| | - Rehan M Hussain
- a Department of Ophthalmology , Indiana University School of Medicine , Indianapolis , IN , USA
| | - Nuria Morral
- c Department of Medical and Molecular Genetics , Indiana University School of Medicine , Indianapolis , IN , USA
| | - Thomas A Ciulla
- a Department of Ophthalmology , Indiana University School of Medicine , Indianapolis , IN , USA.,b Retina Service , Midwest Eye Institute , Indianapolis , IN , USA
| |
Collapse
|
231
|
Weng S, Mao L, Gong Y, Sun T, Gu Q. Role of quercetin in protecting ARPE‑19 cells against H2O2‑induced injury via nuclear factor erythroid 2 like 2 pathway activation and endoplasmic reticulum stress inhibition. Mol Med Rep 2017; 16:3461-3468. [PMID: 28713895 DOI: 10.3892/mmr.2017.6964] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Accepted: 05/18/2017] [Indexed: 11/06/2022] Open
Abstract
Age‑related macular degeneration (AMD) is a common cause of irreversible blindness in the elderly in the western world. Research has demonstrated that degenerative and progressive conditions of retinal pigmented epithelial (RPE) cells are the key pathogenic mechanisms in AMD. Previous research has indicated the anti‑apoptosis, anti‑oxidant, anti‑inflammatory and anti‑cancer properties of quercetin. Therefore, the present study aimed to investigate the protective effects of quercetin on the ARPE‑19 human retinal pigment epithelial cell line. ARPE‑19 cells were pretreated with various concentrations of quercetin (0‑80 µM) before exposure to 300 µM H2O2. Cell viability was assessed and reactive oxygen species (ROS) were determined. The importance of the NF‑E2‑related factor 2 (Nrf2) signaling pathway was corroborated by western blotting and immunostaining. The protein expression levels of endoplasmic reticulum‑associated stress responsive genes and apoptotic markers were assessed by western blotting. The results demonstrated that in the H2O2 group, cell viability was weakened, but preserved in quercetin group in a dose‑dependent manner, particularly at 20 µM. The level of ROS decreased in the quercetin group compared with the control groups. In the quercetin group, the expression levels of Nrf2 and phase II enzymes (NQO1 and HO‑1) were increased, whereas the levels of ER stress markers (binding of immunoglobulin protein, CCAAT/enhancer‑binding protein homologous protein and phosphorylated eukaryotic translation initiation factor 2α) were reduced. Cell apoptosis‑associated protein expression levels were altered, with the increase of B‑cell lymphoma 2 and reduction of Bcl‑2 X‑associated protein. In conclusion, quercetin protected ARPE‑19 cells from H2O2‑induced cytotoxicity by activating the Nrf2 pathway, inhibiting ER stress and targeting anti‑apoptotic proteins.
Collapse
Affiliation(s)
- Sisi Weng
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Hongkou, Shanghai 200080, P.R. China
| | - Lei Mao
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Hongkou, Shanghai 200080, P.R. China
| | - Yuanyuan Gong
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Hongkou, Shanghai 200080, P.R. China
| | - Tao Sun
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Hongkou, Shanghai 200080, P.R. China
| | - Qing Gu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Hongkou, Shanghai 200080, P.R. China
| |
Collapse
|
232
|
Bracha P, Moore NA, Ciulla TA. Induced pluripotent stem cell-based therapy for age-related macular degeneration. Expert Opin Biol Ther 2017; 17:1113-1126. [PMID: 28664762 DOI: 10.1080/14712598.2017.1346079] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION In age-related macular degeneration (AMD), stem cells could possibly replace or regenerate disrupted pathologic retinal pigment epithelium (RPE), and produce supportive growth factors and cytokines such as brain-derived neurotrophic factor. Induced pluripotent stem cells (iPSCs)-derived RPE was first subretinally transplanted in a neovascular AMD patient in 2014. Areas covered: Induced PSCs are derived from the introduction of transcription factors to adult cells under specific cell culture conditions, followed by differentiation into RPE cells. Induced PSC-derived RPE cells exhibit ion transport, membrane potential, polarized VEGF secretion and gene expression that is similar to native RPE. Despite having similar in vitro function, morphology, immunostaining and microscopic analysis, it remains to be seen if iPSC-derived RPE can replicate the myriad of in vivo functions, including immunomodulatory effects, of native RPE cells. Historically, adjuvant RPE transplantation during CNV resections were technically difficult and complicated by immune rejection. Autologous iPSCs are hypothesized to reduce the risk of immune rejection, but their production is time-consuming and expensive. Alternatively, allogenic transplantation using human leukocyte antigen (HLA)-matched iPSCs, similar to HLA-matched organ transplantation, is currently being investigated. Expert opinion: Challenges to successful transplantation with iPSCs include surgical technique, a pathologic subretinal microenvironment, possible immune rejection, and complications of immunosuppression.
Collapse
Affiliation(s)
- Peter Bracha
- a Glick Eye Institute, Department of Ophthalmology , Indiana University School of Medicine , Indianapolis , IN , USA
| | - Nicholas A Moore
- a Glick Eye Institute, Department of Ophthalmology , Indiana University School of Medicine , Indianapolis , IN , USA
| | - Thomas A Ciulla
- a Glick Eye Institute, Department of Ophthalmology , Indiana University School of Medicine , Indianapolis , IN , USA.,b Retina Service , Midwest Eye Institute , Indianapolis , IN , USA
| |
Collapse
|
233
|
Yaspan BL, Williams DF, Holz FG, Regillo CD, Li Z, Dressen A, van Lookeren Campagne M, Le KN, Graham RR, Beres T, Bhangale TR, Honigberg LA, Smith A, Henry EC, Ho C, Strauss EC. Targeting factor D of the alternative complement pathway reduces geographic atrophy progression secondary to age-related macular degeneration. Sci Transl Med 2017. [PMID: 28637922 DOI: 10.1126/scitranslmed.aaf1443] [Citation(s) in RCA: 148] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
| | | | - Frank G Holz
- Department of Ophthalmology, University of Bonn, Bonn, Germany
| | | | - Zhengrong Li
- Genentech Inc., South San Francisco, CA 94080, USA
| | - Amy Dressen
- Genentech Inc., South San Francisco, CA 94080, USA
| | | | - Kha N Le
- Genentech Inc., South San Francisco, CA 94080, USA
| | | | | | | | | | - Ashley Smith
- Genentech Inc., South San Francisco, CA 94080, USA
| | - Erin C Henry
- Genentech Inc., South San Francisco, CA 94080, USA
| | - Carole Ho
- Genentech Inc., South San Francisco, CA 94080, USA
| | | |
Collapse
|
234
|
Koenig P, Sanowar S, Lee CV, Fuh G. Tuning the specificity of a Two-in-One Fab against three angiogenic antigens by fully utilizing the information of deep mutational scanning. MAbs 2017; 9:959-967. [PMID: 28585908 PMCID: PMC5540083 DOI: 10.1080/19420862.2017.1337618] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 05/24/2017] [Accepted: 05/27/2017] [Indexed: 10/19/2022] Open
Abstract
Monoclonal antibodies developed for therapeutic or diagnostic purposes need to demonstrate highly defined binding specificity profiles. Engineering of an antibody to enhance or reduce binding to related antigens is often needed to achieve the desired biologic activity without safety concern. Here, we describe a deep sequencing-aided engineering strategy to fine-tune the specificity of an angiopoietin-2 (Ang2)/vascular endothelial growth factor (VEGF) dual action Fab, 5A12.1 for the treatment of age-related macular degeneration. This antibody utilizes overlapping complementarity-determining region (CDR) sites for dual Ang2/VEGF interaction with KD in the sub-nanomolar range. However, it also exhibits significant (KD of 4 nM) binding to angiopoietin-1, which has high sequence identity with Ang2. We generated a large phage-displayed library of 5A12.1 Fab variants with all possible single mutations in the 6 CDRs. By tracking the change of prevalence of each mutation during various selection conditions, we identified 35 mutations predicted to decrease the affinity for Ang1 while maintaining the affinity for Ang2 and VEGF. We confirmed the specificity profiles for 25 of these single mutations as Fab protein. Structural analysis showed that some of the Fab mutations cluster near a potential Ang1/2 epitope residue that differs in the 2 proteins, while others are up to 15 Å away from the antigen-binding site and likely influence the binding interaction remotely. The approach presented here provides a robust and efficient method for specificity engineering that does not require prior knowledge of the antigen antibody interaction and can be broadly applied to antibody specificity engineering projects.
Collapse
Affiliation(s)
- Patrick Koenig
- Department of Antibody Engineering, Genentech Inc., South San Francisco, CA, USA
| | - Sarah Sanowar
- Department of Antibody Engineering, Genentech Inc., South San Francisco, CA, USA
| | - Chingwei V. Lee
- Department of Antibody Engineering, Genentech Inc., South San Francisco, CA, USA
| | - Germaine Fuh
- Department of Antibody Engineering, Genentech Inc., South San Francisco, CA, USA
| |
Collapse
|
235
|
The novel cyclophilin D inhibitor compound 19 protects retinal pigment epithelium cells and retinal ganglion cells from UV radiation. Biochem Biophys Res Commun 2017; 487:807-812. [DOI: 10.1016/j.bbrc.2017.04.128] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 04/23/2017] [Indexed: 12/23/2022]
|
236
|
Rotenstreich Y, Tzameret A, Kalish SE, Bubis E, Belkin M, Moroz I, Rosner M, Levy I, Margel S, Sher I. A minimally invasive adjustable-depth blunt injector for delivery of pharmaceuticals into the posterior pole. Acta Ophthalmol 2017; 95:e197-e205. [PMID: 27778476 DOI: 10.1111/aos.13238] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 07/25/2016] [Indexed: 12/17/2022]
Abstract
PURPOSE To investigate the feasibility and safety of a novel minimally invasive adjustable-depth blunt injector for pharmaceuticals delivery into the posterior segment. METHODS Indocyanine green (ICG), sodium fluorescein and iron oxide nanoparticles (IONPs) were injected using the new injector into the extravascular spaces of the choroid (EVSC) compartment of rabbits and cadaver pig eyes. Spectral domain optical coherence tomography (SD-OCT), fundus imaging and histology analysis were performed for assessment of injection safety and efficacy. RESULTS Indocyanine green, fluorescein and IONPs were detected across the EVSC in rabbit eyes, covering over 80 per cent of the posterior eye surface. Injected IONPs were retained in the EVSC for at least 2 weeks following injection. No retinal detachment, choroidal haemorrhage or inflammation was detected in any of the injected eyes. In cadaver pig eyes, ICG was detected across the EVSC. CONCLUSIONS This novel minimally invasive delivery system may be used to safely deliver large volumes of pharmaceuticals into a new treatment reservoir compartment - the EVSC which can serve as a depot, in close proximity to the retina, covering most of the surface of the back of the eye without insertion of surgical instruments under the central retina. This system is predicted to enhance the therapeutic effect of treatments for posterior eye disorders.
Collapse
Affiliation(s)
- Ygal Rotenstreich
- Sheba Medical Center; The Maurice and Gabriela Goldschleger Eye Institute; Tel Hashomer Israel
- The Sackler School of Medicine; Tel Aviv University; Tel Aviv Israel
| | - Adi Tzameret
- Sheba Medical Center; The Maurice and Gabriela Goldschleger Eye Institute; Tel Hashomer Israel
- The Sackler School of Medicine; Tel Aviv University; Tel Aviv Israel
| | - Sapir E. Kalish
- Sheba Medical Center; The Maurice and Gabriela Goldschleger Eye Institute; Tel Hashomer Israel
- The Sackler School of Medicine; Tel Aviv University; Tel Aviv Israel
| | - Ettel Bubis
- Sheba Medical Center; The Maurice and Gabriela Goldschleger Eye Institute; Tel Hashomer Israel
- The Sackler School of Medicine; Tel Aviv University; Tel Aviv Israel
| | - Michael Belkin
- Sheba Medical Center; The Maurice and Gabriela Goldschleger Eye Institute; Tel Hashomer Israel
- The Sackler School of Medicine; Tel Aviv University; Tel Aviv Israel
| | - Iris Moroz
- Sheba Medical Center; The Maurice and Gabriela Goldschleger Eye Institute; Tel Hashomer Israel
| | - Mordechai Rosner
- Sheba Medical Center; The Maurice and Gabriela Goldschleger Eye Institute; Tel Hashomer Israel
- The Sackler School of Medicine; Tel Aviv University; Tel Aviv Israel
| | - Itay Levy
- Department of Chemistry; Bar-Ilan Institute of Nanotechnology and Advanced Materials; Ramat-Gan Israel
| | - Shlomo Margel
- Department of Chemistry; Bar-Ilan Institute of Nanotechnology and Advanced Materials; Ramat-Gan Israel
| | - Ifat Sher
- Sheba Medical Center; The Maurice and Gabriela Goldschleger Eye Institute; Tel Hashomer Israel
- The Sackler School of Medicine; Tel Aviv University; Tel Aviv Israel
| |
Collapse
|
237
|
Autophagy regulates Endothelial-Mesenchymal transition by decreasing the phosphorylation level of Smad3. Biochem Biophys Res Commun 2017; 487:740-747. [PMID: 28450107 DOI: 10.1016/j.bbrc.2017.04.130] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 04/23/2017] [Indexed: 12/18/2022]
Abstract
Transforming growth factor-beta2 (TGF-β2) induces Endothelial-Mesenchymal transition (EndoMT) and autophagy in a variety of cells. Previous studies have indicated that activation of autophagy might decrease TGF-β2 induced EndoMT. However, the precise role remains unclear. In the present study, we found that TGF-β2 could induce EndoMT and autophagy in human retinal microvascular endothelial cells (hRMECs). Activation of autophagy by Rapamycin or Trehalose could reduce the expression of Snail, demonstrating a role of autophagy in regulating Snail production both by transcriptional and post-transcriptional mechanism. Co-immunoprecipitation (CoIP) demonstrated that LC3 co-immunoprecipitated with Smad3 and western blot showed that autophagy inducers, Rapamycin and Trehalose, could decrease the phosphorylation level of Smad3. Therefore, our results demonstrate that autophagy counteracts the EndoMT process triggered by TGF-β2 by decreasing the phosphorylation level of Smad3.
Collapse
|
238
|
Li Y, Zhu P, Verma A, Prasad T, Deng H, Yu D, Li Q. A novel bispecific molecule delivered by recombinant AAV2 suppresses ocular inflammation and choroidal neovascularization. J Cell Mol Med 2017; 21:1555-1571. [PMID: 28332318 PMCID: PMC5543459 DOI: 10.1111/jcmm.13086] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 12/13/2016] [Indexed: 02/05/2023] Open
Abstract
Elevated vascular endothelial growth factor (VEGF) and complement activation are implicated in the pathogenesis of different ocular diseases. The objective of this study was to investigate the hypothesis that dual inhibition of both VEGF and complement activation would confer better protection against ocular inflammation and neovascularization. In this study, we engineered a secreted chimeric VEGF inhibitor domain (VID), a complement inhibitor domain (CID) and a dual inhibitor (ACVP1). Vectors expressing these three inhibitors were constructed and packaged into AAV2 (sextY-F) particles. The expression and secretion of the proteins were validated by Western blot. The effects of these inhibitors expressed from AAV2 vectors were examined in endotoxin-induced uveitis (EIU), experimental autoimmune uveoretinitis (EAU) and choroidal neovascularization (CNV) mouse models. The AAV2 vectors expressing the CID- and ACVP1-attenuated inflammation in EIU and EAU model, whereas the vector expressing VID showed improved retinal structure damaged by EAU, but not affect the infiltration of inflammatory cells in EAU or EIU eyes. Both VID and CID vectors improved laser-induced retinal and choroid/RPE injuries and CNV, whereas ACVP1 vector provided significantly better protection. Our results suggest that gene therapy targeting VEGF and complement components could provide an innovative and long-term strategy for ocular inflammatory and neovascular diseases.
Collapse
Affiliation(s)
- Yiming Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, China.,Department of Ophthalmology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Ping Zhu
- Department of Ophthalmology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Amrisha Verma
- Department of Ophthalmology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Tuhina Prasad
- Department of Ophthalmology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Hongxin Deng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, China
| | - Dechao Yu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, China
| | - Qiuhong Li
- Department of Ophthalmology, College of Medicine, University of Florida, Gainesville, FL, USA
| |
Collapse
|
239
|
Qi X, Pay SL, Yan Y, Thomas J, Lewin AS, Chang LJ, Grant MB, Boulton ME. Systemic Injection of RPE65-Programmed Bone Marrow-Derived Cells Prevents Progression of Chronic Retinal Degeneration. Mol Ther 2017; 25:917-927. [PMID: 28202390 PMCID: PMC5383551 DOI: 10.1016/j.ymthe.2017.01.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 01/04/2017] [Accepted: 01/06/2017] [Indexed: 01/18/2023] Open
Abstract
Bone marrow stem and progenitor cells can differentiate into a range of non-hematopoietic cell types, including retinal pigment epithelium (RPE)-like cells. In this study, we programmed bone marrow-derived cells (BMDCs) ex vivo by inserting a stable RPE65 transgene using a lentiviral vector. We tested the efficacy of systemically administered RPE65-programmed BMDCs to prevent visual loss in the superoxide dismutase 2 knockdown (Sod2 KD) mouse model of age-related macular degeneration. Here, we present evidence that these RPE65-programmed BMDCs are recruited to the subretinal space, where they repopulate the RPE layer, preserve the photoreceptor layer, retain the thickness of the neural retina, reduce lipofuscin granule formation, and suppress microgliosis. Importantly, electroretinography and optokinetic response tests confirmed that visual function was significantly improved. Mice treated with non-modified BMDCs or BMDCs pre-programmed with LacZ did not exhibit significant improvement in visual deficit. RPE65-BMDC administration was most effective in early disease, when visual function and retinal morphology returned to near normal, and less effective in late-stage disease. This experimental paradigm offers a minimally invasive cellular therapy that can be given systemically overcoming the need for invasive ocular surgery and offering the potential to arrest progression in early AMD and other RPE-based diseases.
Collapse
Affiliation(s)
- Xiaoping Qi
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - S Louise Pay
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Yuanqing Yan
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL 32610, USA
| | - James Thomas
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL 32610, USA
| | - Alfred S Lewin
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL 32610, USA
| | - Lung-Ji Chang
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL 32610, USA
| | - Maria B Grant
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Michael E Boulton
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
240
|
Simple and complex retinal dystrophies are associated with profoundly different disease networks. Sci Rep 2017; 7:41835. [PMID: 28139756 PMCID: PMC5282568 DOI: 10.1038/srep41835] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 12/28/2016] [Indexed: 12/20/2022] Open
Abstract
Retinopathies are a group of monogenetic or complex retinal diseases associated with high unmet medical need. Monogenic disorders are caused by rare genetic variation and usually arise early in life. Other diseases, such as age-related macular degeneration (AMD), develop late in life and are considered to be of complex origin as they develop from a combination of genetic, ageing, environmental and lifestyle risk factors. Here, we contrast the underlying disease networks and pathological mechanisms of monogenic as opposed to complex retinopathies, using AMD as an example of the latter. We show that, surprisingly, genes associated with the different forms of retinopathies in general do not overlap despite their overlapping retinal phenotypes. Further, AMD risk genes participate in multiple networks with interaction partners that link to different ubiquitous pathways affecting general tissue integrity and homeostasis. Thus AMD most likely represents an endophenotype with differing underlying pathogenesis in different subjects. Localising these pathomechanisms and processes within and across different retinal anatomical compartments provides a novel representation of AMD that may be extended to complex disease in general. This approach may generate improved treatment options that target multiple processes with the aim of restoring tissue homeostasis and maintaining vision.
Collapse
|
241
|
Feltgen N, Bertelmann T, Bretag M, Pfeiffer S, Hilgers R, Callizo J, Goldammer L, Bemme S, Hoerauf H. Efficacy and safety of a fixed bimonthly ranibizumab treatment regimen in eyes with neovascular age-related macular degeneration: results from the RABIMO trial. Graefes Arch Clin Exp Ophthalmol 2017; 255:923-934. [PMID: 28102456 DOI: 10.1007/s00417-017-3589-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 01/05/2017] [Accepted: 01/11/2017] [Indexed: 10/20/2022] Open
Abstract
PURPOSE To evaluate prospectively the efficacy and safety of a fixed bimonthly ranibizumab treatment regimen (RABIMO) in eyes with neovascular age-related macular degeneration (nAMD) and to compare these results with a pro re nata (PRN) treatment scheme. METHODS This was a 12-month, phase IV, single center, randomised, non-inferiority study. Following three initial monthly injections, patients were randomised to receive either ranibizumab bimonthly (RABIMO group) or ranibizumab PRN (PRN group) (n = 20 each). Main outcome measures were best-corrected visual acuity (BCVA), central retinal thickness (CRT), number of injections, and adverse events (AEs). RESULTS BCVA [median (interquartile range, IQR)] increased significantly in both groups after 12 months [RABIMO group +8.5 (14); PRN group +6.5 (16) ETDRS letters] when compared to baseline (p < 0.0001; p = 0.0085). At month 12, the RABIMO treatment regimen was non-inferior to the PRN scheme (∆BCVA = 3.5 ETDRS letters; p < 0.0001). CRT was significantly reduced in both groups after the 12-month study period (p < 0.0001 each), with no significant difference between groups (p = 0.6772). Number of overall injections [median (IQR)] was 8 (0) in the RABIMO versus 4 (5) in the PRN group (p = 0.0037). Three patients in the RABIMO group received one additional unscheduled injection. We observed no significant differences between groups in the number of patients with reported SAEs/AEs (RABIMO group n = 6/15; PRN group n = 7/13) (p = 0.7357/p = 0.4902). CONCLUSIONS We found no evidence of significant functional or anatomical differences between the RABIMO and PRN treatment regimens. However, the RABIMO group's number of injections was twice as high as the PRN group's (protocol-driven). In light of potential side effects, the fixed bimonthly treatment regimen might not be advisable for routine clinical care, but it might be a worthwhile treatment option if monthly monitoring is not possible. Eudra-CT number: 2009-017324-11.
Collapse
Affiliation(s)
- Nicolas Feltgen
- Department of Ophthalmology, University Medical Centre, Robert-Koch-Straße 40, 37075, Goettingen, Germany.
| | - Thomas Bertelmann
- Department of Ophthalmology, University Medical Centre, Robert-Koch-Straße 40, 37075, Goettingen, Germany
| | - Mirko Bretag
- Department of Ophthalmology, Carl-Thiem-Klinikum gGmbH, Cottbus, Germany
| | - Sebastian Pfeiffer
- Institute for Clinical Research GmbH, Georg-August-University, Goettingen, Germany
| | - Reinhard Hilgers
- Institute for Medical Statistics, Georg-August-University, Goettingen, Germany
| | - Josep Callizo
- Department of Ophthalmology, University Medical Centre, Robert-Koch-Straße 40, 37075, Goettingen, Germany
| | | | - Sebastian Bemme
- Department of Ophthalmology, University Medical Centre, Robert-Koch-Straße 40, 37075, Goettingen, Germany
| | - Hans Hoerauf
- Department of Ophthalmology, University Medical Centre, Robert-Koch-Straße 40, 37075, Goettingen, Germany
| |
Collapse
|
242
|
Toledano S, Lu H, Palacio A, Ziv K, Kessler O, Schaal S, Neufeld G, Barak Y. A Sema3C Mutant Resistant to Cleavage by Furin (FR-Sema3C) Inhibits Choroidal Neovascularization. PLoS One 2016; 11:e0168122. [PMID: 28036336 PMCID: PMC5201251 DOI: 10.1371/journal.pone.0168122] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 11/23/2016] [Indexed: 01/13/2023] Open
Abstract
In age-related macular degeneration (AMD), abnormal sub retinal choroidal neovascularization (CNV) is a major cause of blindness. FR-sema3C is a point mutated form of semaphorin-3C that is resistant to cleavage by furin like pro-protein convertases (FPPC). We have found in previous work that FR-sema3C functions as an anti-angiogenic factor. In this study we investigated the possible use of FR-sema3C as an inhibitor of CNV. FR-sema3C inhibits VEGF as well as PDGF-BB signal transduction in endothelial cells and to less extent bFGF induced signal transduction using a mechanism that does not depend upon the binding of VEGF like the drugs that are currently the mainstay treatment for AMD. CNV was induced in eyes of C57 black mice by laser photocoagulation. Intravitreal injection of FR-Sema3C or aflibercept (VEGF-trap) was then used to inhibit CNV formation. Invading choroidal vessels were visualized a week later by injection of FITC-dextran into the circulation, followed by the measurement of the area of the invading blood vessels. Injection of 0.1 μg FR-Sema3C inhibited CNV by 55% (P<0.01) and was as effective as 5 μg aflibercept. FR-sema3C did not display any adverse effects on retinal function following its injection into eyes of healthy mice as assessed by optokinetic reflex (OKR) and Electro-retinogram (ERG) criteria. Furthermore, FR-sema3C did not induce apoptosis in the retina as determined by TUNEL nor was there any discernable structural damage to the retina as assessed by several immuno-histochemical criteria. Our results suggest that FR-sema3C could perhaps be used for the treatment of AMD, and that it may perhaps be of benefit to patients that do not respond well to current treatments relying on VEGF sequestering agents.
Collapse
Affiliation(s)
- Shira Toledano
- Cancer Research and Vascular Biology Center, The Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| | - Huayi Lu
- Second Hospital of Jilin University, Changchun, Jilin Province, P.R. China
- Department of Ophthalmology and Visual Sciences, University of Louisville, Louisville, Kentucky, United States of America
| | - Agustina Palacio
- Department of Ophthalmology and Visual Sciences, University of Louisville, Louisville, Kentucky, United States of America
| | - Keren Ziv
- Cancer Research and Vascular Biology Center, The Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| | - Ofra Kessler
- Cancer Research and Vascular Biology Center, The Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| | - Shlomit Schaal
- Department of Ophthalmology and Visual Sciences, University of Louisville, Louisville, Kentucky, United States of America
- Department of Ophthalmology and Visual Sciences, University of Massachusetts School of Medicine, Massachusetts, United States of America
- * E-mail: (SS); (GN); (YB)
| | - Gera Neufeld
- Cancer Research and Vascular Biology Center, The Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
- * E-mail: (SS); (GN); (YB)
| | - Yoreh Barak
- Department of Ophthalmology, RAMBAM Medical Center, Haifa, Israel
- * E-mail: (SS); (GN); (YB)
| |
Collapse
|
243
|
Lee BL, Kang JH, Kim HM, Jeong SH, Jang DS, Jang YP, Choung SY. Polyphenol-enriched Vaccinium uliginosum L. fractions reduce retinal damage induced by blue light in A2E-laden ARPE19 cell cultures and mice. Nutr Res 2016; 36:1402-1414. [PMID: 27993192 DOI: 10.1016/j.nutres.2016.11.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 11/15/2016] [Accepted: 11/16/2016] [Indexed: 01/01/2023]
Abstract
Polyphenols exert beneficial effects on vision. We hypothesized that polyphenol components of Vaccinium uliginosum L. (V.U.) extract protect retinal pigment epithelial (RPE) cells against blue light-induced damage. Our aim was to test extracts containing polyphenol components to ascertain effects to reduce damage against blue light in RPEs. We measured the activity in fractions eluted from water, ethanol, and HP20 resin (FH), and found that the FH fraction had the highest beneficial activity. We isolated the individual active compounds from the FH fraction using chromatographic techniques, and found that FH contained flavonoids, anthocyanins, phenyl propanoids, and iridoids. Cell cultures of A2E-laden ARPE-19 exposed to blue light after treatment with V.U. extract fractions and their individual constituents indicated improvement. V uliginosum L extract fractions and constituent compounds significantly reduced A2E photo-oxidation-induced RPE cell death and inhibited intracellular A2E accumulation. Furthermore, Balb/c male mice were exposed to blue light at 10000 lux for 1 h/d for 2 weeks to induce retinal damage. One week after the final blue light exposure, retinal damage evaluated revealed that the outer nuclear layer thickness and nuclei count were improved. Histologic examination of murine photoreceptor cells demonstrated that FH, rich in polyphenols, inhibited the loss of outer nuclear layer thickness and nuclei. Our findings suggest that V.U. extract and eluted fractions are a potential source of bioactive compounds that potentially serve a therapeutic approach for age-related macular degeneration.
Collapse
Affiliation(s)
- Bom-Lee Lee
- Department of Life and Nanopharmaceutical Sciences of Pharmacy, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Jung-Hwan Kang
- Department of Life and Nanopharmaceutical Sciences of Pharmacy, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Hye-Mi Kim
- Department of Preventive Pharmacy and Toxicology, College of Pharmacy, Kyung Hee University, Seoul, Republic of Korea
| | - Se-Hee Jeong
- Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul, Republic of Korea
| | - Dae-Sik Jang
- Department of Life and Nanopharmaceutical Sciences of Pharmacy, Graduate School, Kyung Hee University, Seoul, Republic of Korea; Department of Preventive Pharmacy Science, College of Pharmacy, Kyung Hee University, Seoul, Republic of Korea
| | - Young-Pyo Jang
- Department of Life and Nanopharmaceutical Sciences of Pharmacy, Graduate School, Kyung Hee University, Seoul, Republic of Korea; Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul, Republic of Korea
| | - Se-Young Choung
- Department of Life and Nanopharmaceutical Sciences of Pharmacy, Graduate School, Kyung Hee University, Seoul, Republic of Korea; Department of Preventive Pharmacy and Toxicology, College of Pharmacy, Kyung Hee University, Seoul, Republic of Korea.
| |
Collapse
|
244
|
Du Z, Wu X, Song M, Li P, Wang L. Oxidative damage induces MCP-1 secretion and macrophage aggregation in age-related macular degeneration (AMD). Graefes Arch Clin Exp Ophthalmol 2016; 254:2469-2476. [PMID: 27812755 DOI: 10.1007/s00417-016-3508-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 07/26/2016] [Accepted: 10/03/2016] [Indexed: 01/21/2023] Open
Abstract
PURPOSE Age-related macular degeneration (AMD) is a major cause of progressive and degenerative visual impairment. Although the exact pathogenic mechanism of AMD is still unknown, clinical observations such as the high accumulation of oxidative products and macrophages in retina suggest the importance of oxidative stress and inflammation in AMD. METHODS Mouse photoreceptor-derived 661 W cells and human ARPE-19 cells were treated with oxidized phospholipids (Ox-PC) or H2O2 to mimic oxidative damage. The effect of monocyte chemoattractant protein 1 (MCP-1) secreted by retina cells on the migration of monocyte macrophage RAW 264.7 cells was determined using transwell chambers and antibody neutralization assay. MCP-1, tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and vascular endothelial growth factor (VEGF) that secreted into supernatant were measured by ELISA and their intracellular expression was detected by qRT-PCR and western blot. Intracellular Ox-PC level was detected by competitive ELISA. The amount of migrated RAW 264.7 cells was counted by flow cytometry. RESULTS Oxidative damage by both H2O2 and Ox-PC induced the secretion of MCP-1 in human ARPE-19 and mouse 661 W cells. MCP-1 induced by oxidative damage enhanced the migration ability of macrophage RAW 264.7 cells and the secretion of TNF-α, IL-1β and VEGF, which could be reduced by anti-MCP-1 neutralizing antibodies. CONCLUSION The results indicated that oxidative damage increases intracellular Ox-PC and the secretion of MCP-1 in retina cells. The increased MCP-1 induced by oxidative damage attracts macrophages to retinas, and macrophages release pro-inflammatory factor and promote the process of AMD.
Collapse
Affiliation(s)
- Zhaojiang Du
- Department of Ophthalmology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China.
| | - Xuemei Wu
- Department of Ophthalmology, Chinese Medicine Research Institute, Xi'an, 710003, Shannxi Province, China
| | - Meixia Song
- Department of Ophthalmology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China.,Department of Ophthalmology, 153rd Central Hospital of PLA, Zhengzhou, 450007, Henan Province, China
| | - Peng Li
- Department of Ophthalmology, No.451 Hospital of PLA, Xi'an, 710054, China
| | - Li Wang
- Department of Optometry, Xi'an Medical College, Xi'an, 710021, China
| |
Collapse
|
245
|
Carver KA, Lin CM, Bowes Rickman C, Yang D. Lack of the P2X 7 receptor protects against AMD-like defects and microparticle accumulation in a chronic oxidative stress-induced mouse model of AMD. Biochem Biophys Res Commun 2016; 482:81-86. [PMID: 27810364 DOI: 10.1016/j.bbrc.2016.10.140] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 10/29/2016] [Indexed: 12/17/2022]
Abstract
The P2X7 receptor (P2X7R) is an ATP-gated ion channel that is a key player in oxidative stress under pathological conditions. The P2X7R is expressed in the retinal pigmented epithelium (RPE) and neural retina. Chronic oxidative stress contributes to the pathogenesis of age-related macular degeneration (AMD). Mice lacking Cu, Zn superoxide dismutase (Sod1) developed chronic oxidative stress as well as AMD-like features, but whether the P2X7R plays a causative role in oxidative stress-induced AMD is unknown. Thus, the main purpose of this study was to test if concurrent knockout (KO) of P2X7R could block AMD-like defects seen in Sod1 KO mice. Using multiple approaches, we demonstrate that Sod1 KO causes AMD-like defects, including positive staining for oxidative stress markers, 3-nitrotyrosine and carboxymethyl lysine, thinning of the RPE and retina, thickening of Bruch's membrane, presence of basal laminar and linear deposits, RPE barrier disruption and accumulation of microglia/macrophages. Moreover, we find that Sod1 KO mice accumulate more microparticles (MPs) within RPE/choroid tissues. Concurrent KO of the P2X7R protects against AMD-like defects and MP accumulation in Sod1 KO mice. Together, we show for the first time, that deficiency of P2X7R prevents in vivo oxidative stress-induced accumulation of MPs and AMD-like defects. This work could potentially lead to novel therapies for AMD and other oxidative stress-driven diseases.
Collapse
Affiliation(s)
- Kyle A Carver
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105, USA
| | - C M Lin
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105, USA
| | - Catherine Bowes Rickman
- Department of Ophthalmology, Duke Eye Center, Duke University, Durham, NC 27710, USA; Department of Cell Biology, Duke University, Durham, NC 27710, USA
| | - Dongli Yang
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105, USA.
| |
Collapse
|
246
|
Matsunaga D, Sreekumar PG, Ishikawa K, Terasaki H, Barron E, Cohen P, Kannan R, Hinton DR. Humanin Protects RPE Cells from Endoplasmic Reticulum Stress-Induced Apoptosis by Upregulation of Mitochondrial Glutathione. PLoS One 2016; 11:e0165150. [PMID: 27783653 PMCID: PMC5081188 DOI: 10.1371/journal.pone.0165150] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Accepted: 10/09/2016] [Indexed: 01/31/2023] Open
Abstract
Humanin (HN) is a small mitochondrial-encoded peptide with neuroprotective properties. We have recently shown protection of retinal pigmented epithelium (RPE) cells by HN in oxidative stress; however, the effect of HN on endoplasmic reticulum (ER) stress has not been evaluated in any cell type. Our aim here was to study the effect of HN on ER stress-induced apoptosis in RPE cells with a specific focus on ER-mitochondrial cross-talk. Dose dependent effects of ER stressors (tunicamycin (TM), brefeldin A, and thapsigargin) were studied after 12 hr of treatment in confluent primary human RPE cells with or without 12 hr of HN pretreatment (1-20 μg/mL). All three ER stressors induced RPE cell apoptosis in a dose dependent manner. HN pretreatment significantly decreased the number of apoptotic cells with all three ER stressors in a dose dependent manner. HN pretreatment similarly protected U-251 glioma cells from TM-induced apoptosis in a dose dependent manner. HN pretreatment significantly attenuated activation of caspase 3 and ER stress-specific caspase 4 induced by TM. TM treatment increased mitochondrial superoxide production, and HN co-treatment resulted in a decrease in mitochondrial superoxide compared to TM treatment alone. We further showed that depleted mitochondrial glutathione (GSH) levels induced by TM were restored with HN co-treatment. No significant changes were found for the expression of several antioxidant enzymes between TM and TM plus HN groups except for the expression of glutamylcysteine ligase catalytic subunit (GCLC), the rate limiting enzyme required for GSH biosynthesis, which is upregulated with TM and TM+HN treatment. These results demonstrate that ER stress promotes mitochondrial alterations in RPE that lead to apoptosis. We further show that HN has a protective effect against ER stress-induced apoptosis by restoring mitochondrial GSH. Thus, HN should be further evaluated for its therapeutic potential in disorders linked to ER stress.
Collapse
Affiliation(s)
- Douglas Matsunaga
- Department of Pathology and Ophthalmology, USC Roski Eye Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA, United States of America
| | - Parameswaran G. Sreekumar
- Arnold and Mabel Beckman Macular Research Center, Doheny Eye Institute, Los Angeles, CA, United States of America
| | - Keijiro Ishikawa
- Arnold and Mabel Beckman Macular Research Center, Doheny Eye Institute, Los Angeles, CA, United States of America
| | - Hiroto Terasaki
- Arnold and Mabel Beckman Macular Research Center, Doheny Eye Institute, Los Angeles, CA, United States of America
| | - Ernesto Barron
- Arnold and Mabel Beckman Macular Research Center, Doheny Eye Institute, Los Angeles, CA, United States of America
| | - Pinchas Cohen
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, United States of America
| | - Ram Kannan
- Arnold and Mabel Beckman Macular Research Center, Doheny Eye Institute, Los Angeles, CA, United States of America
| | - David R. Hinton
- Department of Pathology and Ophthalmology, USC Roski Eye Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA, United States of America
| |
Collapse
|
247
|
Toledano S, Lu H, Palacio A, Kigel B, Kessler O, Allon G, Barak Y, Neufeld G, Schaal S. A SEMA3E mutant resistant to cleavage by furins (UNCL-SEMA3E) inhibits choroidal neovascularization. Exp Eye Res 2016; 153:186-194. [PMID: 27725196 DOI: 10.1016/j.exer.2016.10.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 08/30/2016] [Accepted: 10/05/2016] [Indexed: 01/16/2023]
Abstract
Abnormal subretinal choroidal neovascularization (CNV) is a major cause of blindness in exudative age-related macular degeneration (AMD). Current anti-angiogenic treatments by VEGF sequestering agents have been successful, but a significant proportion of patients do not respond well to these treatments, and the response of others diminishes over time, suggesting that additional anti-angiogenic agents that function by separate mechanisms may be of use to such patients. We have previously found that a point mutated form of semaphorin-3E resistant to cleavage by furin like pro-protein convertases (UNCL-Sema3E) displays potent anti-angiogenic properties. We therefore determined if UNCL-Sema3E has potential as an inhibitor of CNV formation. We chose to study UNCL-Sema3E rather than wild type sema3E because unlike full length sema3E, the major p61-Sema3E peptide that is produced by cleavage of sema3E with furin like pro-protein convertases activates signal transduction mediated by the ErbB2 receptor and can promote tumor metastasis in addition to its anti-angiogenic activity. UNCL-Sema3E inhibited efficiently vascular endothelial growth factor-A (VEGF), platelet derived growth factor (PDGF) and basic fibroblast growth factor (bFGF) signaling in human umbilical vein derived endothelial cells (HUVEC) and to a lesser extent hepatocyte growth factor (HGF) signal transduction. CNV that was induced in the eyes of C57 black mice by laser photocoagulation was inhibited by 65% (P < 0.01) following a single bolus intra-vitreal injection of 5 μg UNCL-Sema3E. This inhibitory effect was similar to the inhibition produced by a single bolus intra-vitreal injection of 5 μg aflibercept. A similar inhibition of CNV was observed following the injection of UNCL-Sema3E into the eyes of Long-Evans rats. However, a higher dose of UNCL-Sema3E (125 μg), partially due to the larger volume of the vitreous cavity of rats, was required to achieve maximal inhibition of CNV. Injection of UNCL-Sema3E into eyes of healthy mice did not have any adverse effect on retinal function as assessed by optic kinetic reflex (OKR) or by electroretinogram (ERG) assays nor did UNCL-Sema3E injection affect the structure of the retina as determined using histology. To conclude, our results suggest that UNCL-Sema3E may be useful for the treatment of exudative AMD, which does not respond well to conventional anti-VEGF therapy.
Collapse
Affiliation(s)
- Shira Toledano
- Cancer Research and Vascular Biology Center, The Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| | - Huayi Lu
- Second Hospital of Jilin University, Changchun, Jilin Province, 130041, PR China; Department of Ophthalmology and Visual Sciences, University of Louisville, KY, USA
| | - Agustina Palacio
- Department of Ophthalmology and Visual Sciences, University of Louisville, KY, USA
| | - Boaz Kigel
- Cancer Research and Vascular Biology Center, The Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| | - Ofra Kessler
- Cancer Research and Vascular Biology Center, The Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| | - Gilad Allon
- Department of Ophthalmology, Rambam Medical Center, Haifa, Israel
| | - Yoreh Barak
- Department of Ophthalmology, Rambam Medical Center, Haifa, Israel.
| | - Gera Neufeld
- Cancer Research and Vascular Biology Center, The Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel.
| | - Shlomit Schaal
- Department of Ophthalmology and Visual Sciences, University of Louisville, KY, USA; Department of Ophthalmology and Visual Sciences, University of Massachusetts School of Medicine, USA.
| |
Collapse
|
248
|
Nuclear Factor (Erythroid-Derived)-Related Factor 2-Associated Retinal Pigment Epithelial Cell Protection under Blue Light-Induced Oxidative Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:8694641. [PMID: 27774118 PMCID: PMC5059614 DOI: 10.1155/2016/8694641] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Revised: 07/29/2016] [Accepted: 08/25/2016] [Indexed: 12/18/2022]
Abstract
Purpose. It is a matter of increasing concern that exposure to light-emitting diodes (LED), particularly blue light (BL), damages retinal cells. This study aimed to investigate the retinal pigment epithelium (RPE) damage caused by BL and to elucidate the role of nuclear factor (erythroid-derived)-related factor 2 (Nrf2) in the pathogenesis of BL-induced RPE damage. Methods. ARPE-19, a human RPE cell line, and mouse primary RPE cells from wild-type and Nrf2 knockout (Nrf2-/-) mice were cultured under blue LED exposure (intermediate wavelength, 450 nm). Cell death rate and reactive oxygen species (ROS) generation were measured. TUNEL staining was performed to detect apoptosis. Real-time polymerase chain reaction was performed on NRF2 mRNA, and western blotting was performed to detect Nrf2 proteins in the nucleus or cytoplasm of RPE cells. Results. BL exposure increased cell death rate and ROS generation in ARPE-19 cells in a time-dependent manner; cell death was caused by apoptosis. Moreover, BL exposure induced NRF2 mRNA upregulation and Nrf2 nuclear translocation in RPE. Cell death rate was significantly higher in RPE cells from Nrf2-/- mice than from wild-type mice. Conclusions. The Nrf2 pathway plays an important role in protecting RPE cells against BL-induced oxidative stress.
Collapse
|
249
|
Kang JH, Choung SY. Protective effects of resveratrol and its analogs on age-related macular degeneration in vitro. Arch Pharm Res 2016; 39:1703-1715. [PMID: 27659166 DOI: 10.1007/s12272-016-0839-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 09/11/2016] [Indexed: 12/13/2022]
Abstract
Damage of retinal pigment epithelial (RPE) cells by A2E may be critical for age-related macular degeneration (AMD) management. Accumulation and photooxidation of A2E are known to be one of the critical causes in AMD. Here, we evaluated the protective effect of resveratrol (RES), piceatannol (PIC) and RES glycones on blue-light-induced RPE cell death caused by A2E photooxidation. A2E treatment followed by blue light exposure caused significant damages on human RPE cells (ARPE-19). But the damages were attenuated by post- and pre-treatment of RES and PIC in our in vitro models. The results of cell free system and FAB-MS analysis clearly showed that the reduction of A2E by blue light exposure was significantly rescued, and that oxidized forms of A2E were significantly reduced by RES or PIC treatment. Besides, RES or PIC inhibited the intracellular accumulation of A2E. Not only RES and PIC but RES glycones showed protection of ARPE-19 cells against A2E and blue-light-induced photo-damage. These findings demonstrate that RES and its analogs may have protective effects against A2E and blue-light-induced ARPE-19 cell death through regulation of A2E accumulation as well as photooxidation of A2E. Thus RES and its analogs may be beneficial for AMD treatment.
Collapse
Affiliation(s)
- Jung-Hwan Kang
- Department of Life and Nanopharmaceutical Science of Pharmacy, College of Pharmacy, Kyung Hee University, Seoul, Republic of Korea
| | - Se-Young Choung
- Department of Life and Nanopharmaceutical Science of Pharmacy, College of Pharmacy, Kyung Hee University, Seoul, Republic of Korea. .,Department of Preventive Pharmacy and Toxicology, College of Pharmacy, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea.
| |
Collapse
|
250
|
Garweg JG. Makula-Atrophie bei feuchter altersabhängiger Makuladegeneration. Ophthalmologe 2016; 113:1036-1045. [DOI: 10.1007/s00347-016-0306-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|