1
|
Gharegezloo Z, Rezvani Z, Sanie-Jahromi F, Namjoyan F. The effect of Coix lachrymal L. seed extract on the expression of inflammation and fibrogenesis genes in human retinal pigment epithelial cells. Biomed Pharmacother 2024; 181:117646. [PMID: 39486365 DOI: 10.1016/j.biopha.2024.117646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 10/16/2024] [Accepted: 10/28/2024] [Indexed: 11/04/2024] Open
Abstract
Proliferative vitreoretinopathy (PVR) is a vision-threatening condition associated with retinal-detachment (RD), primarily caused by fibrocellular scar membrane formation. This study investigates the therapeutic potential of adlay seed extract fractions in mitigating PVR-associated pathways, focusing on oxidative stress, proliferation, inflammation, and fibrogenesis in retinal pigment epithelial (RPE) cells. Adlay seed extract fractions (methanolic: MeOH and residual: Res) were obtained through solvent extraction and characterized for carbohydrate, protein, flavonoid content, and antioxidant activity. RPE cells were cultured, and their viability in response to adlay fractions was assessed using the MTT assay. Gene expression analysis of IL-1β, IL-6, LIF, TGF-β, Snail and α-SMA genes was conducted via real-time PCR after treatment with adlay fractions. The Res fraction exhibited higher levels of protein, carbohydrate, flavonoids, and phenols compared to the MeOH fraction, along with significantly enhanced antioxidant activity. Both fractions showed inhibitory effects on RPE cell viability, with the Res fraction demonstrating a more pronounced impact. Gene expression analysis revealed a significant decrease in IL-6 and TGF-β expression with the MeOH fraction treatment, while the Res fraction led to decreased expression of IL-6, LIF, TGF-β, Snail and α-SMA, indicating a more comprehensive modulation of PVR-associated pathways. This study highlights the potential therapeutic benefits of adlay seed extract fractions in mitigating PVR-associated pathways in RPE cells. The Res fraction, particularly rich in bioactive compounds and exhibiting potent antioxidant activity, shows promise in attenuating oxidative stress, proliferation, inflammation, and fibrogenesis, critical processes in PVR development. These findings underscore the potential of adlay seed extracts as a novel therapeutic strategy for PVR warranting further investigation and clinical validation.
Collapse
Affiliation(s)
- Zahra Gharegezloo
- Division of biotechnology, Department of Cell and Molecular Biology, Faculty of Chemistry, University of Kashan, Kashan, Iran.
| | - Zahra Rezvani
- Division of biotechnology, Department of Cell and Molecular Biology, Faculty of Chemistry, University of Kashan, Kashan, Iran.
| | - Fatemeh Sanie-Jahromi
- Poostchi Ophthalmology Research Center, Department of Ophthalmology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Foroogh Namjoyan
- Pharmacognosy Department, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
2
|
Mysore Y, Hytti M, Deen AJ, Ranta-Aho S, Piippo N, Toppila M, Loukovaara S, Harju N, Kauppinen A. Epithelial-mesenchymal Transition (EMT) and the Effect of Atorvastatin on it in ARPE-19 cells. Cell Biochem Biophys 2024; 82:1523-1536. [PMID: 38777991 PMCID: PMC11344705 DOI: 10.1007/s12013-024-01305-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/07/2024] [Indexed: 05/25/2024]
Abstract
Proliferative vitreoretinopathy (PVR) develops after an unsuccessful or complicated recovery from rhegmatogenous retinal detachment (RRD) surgery. Intraocular scar formation with the contribution of epithelial-mesenchymal transition (EMT) in RPE cells is prominent in the pathology of PVR. In the present study, the EMT process was experimentally induced in human retinal pigment epithelium (RPE; ARPE-19) cells, and the effect of atorvastatin on the process was studied. The mRNA and protein levels of mesenchymal markers actin alpha 2 (ACTA2) / alpha-smooth muscle actin (α-SMA) and fibronectin (FN), and epithelial markers occludin (OCLN) and zonula occludens-1 (ZO-1) were measured using quantitative real-time PCR (qRT-PCR) and western blot methods, respectively. In addition, α-SMA and FN were visualized using immunofluorescence staining. Cells were photographed under a phase contrast light microscope. Changes in the functionality of cells following the EMT process were studied using the IncuCyte scratch wound cell migration assay and the collagen cell invasion assay with confocal microscopy. The induction of EMT in ARPE-19 cells increased the expression of mesenchymal markers ACTA2/α-SMA and fibronectin and reduced the expression of epithelial marker OCLN both at mRNA and protein levels. The mRNA levels of ZO-1 were lower after EMT, as well. Increased levels of α-SMA and FN were confirmed by immunofluorescence staining. Atorvastatin further increased the mRNA levels of mesenchymal markers ACTA2 and FN as well as the protein levels of α-SMA and reduced the mRNA levels of epithelial markers OCLN and ZO-1 under the EMT process. EMT promoted wound closure and cell invasion into the 3D collagen matrix when compared to untreated control cells. These data present cellular changes upon the induction of the EMT process in ARPE-19 cells and the propensity of atorvastatin to complement the effect. More studies are needed to confirm the exact influence of the EMT process and atorvastatin treatment on the PVR development after RRD surgery.
Collapse
Affiliation(s)
- Yashavanthi Mysore
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Maria Hytti
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
- Department of Ophthalmology, Kuopio University Hospital and School of Medicine, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Ashik Jawahar Deen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Sofia Ranta-Aho
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Niina Piippo
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Maija Toppila
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Sirpa Loukovaara
- Department of Ophthalmology, Unit of Vitreoretinal Surgery, Helsinki University Central Hospital, and Individualized Drug Therapy Research Program, University of Helsinki, Helsinki, Finland
| | - Niina Harju
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland.
- Head and Neck Center, Ophthalmology Research Unit, Helsinki University Central Hospital, Helsinki, Finland.
| | - Anu Kauppinen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland.
| |
Collapse
|
3
|
Hao X, Hua Y, Xie C, Xu H. MiR-21 Participates in Anti-VEGF-Induced Epithelial Mesenchymal Transformation in RPE Cells. Clin Ophthalmol 2023; 17:3047-3056. [PMID: 37869043 PMCID: PMC10588660 DOI: 10.2147/opth.s427894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Accepted: 10/05/2023] [Indexed: 10/24/2023] Open
Abstract
Background To explore the role and possible mechanism of miR-21 in anti-VEGF drug-induced epithelial-mesenchymal transformation (EMT) in human retinal pigment epithelium (ARPE-19) cells, and to seek more therapeutic targets to improve prognosis vision. Methods ARPE-19 cells were exposed to clinical dosage of bevacizumab and miR-21 expression was measured by real-time polymerase chain reaction (RT-PCR) assay. MiR-21 mimic and inhibitor were transfected into bevacizumab-induced ARPE-19, the expression of α-smooth muscle actin (α-SMA), E-cadherin, and SNAI1 were detected by cell immunofluorescence and Western blotting. Results Clinical dosage of bevacizumab caused EMT and enhanced miR-21 expression in ARPE-19 cells (P<0.05). The inhibition of miR-21 attenuated the EMT effect of bevacizumab, while overexpression of miR-21 promoted this activity (P<0.05). The SNAI1 was up-regulated by bevacizumab and promotion was partially suppressed by the miR-21 inhibitor and aggravated by the miR-21 mimic (P<0.05). Conclusion MiR-21 promotes bevacizumab-induced EMT in ARPE cells which is significantly positively correlated with SNAI1. MiR-21 might be a potential miRNA-based therapeutic target for reducing bevacizumab-induced subretinal fibrosis.
Collapse
Affiliation(s)
- Xianghui Hao
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao, People’s Republic of China
| | - Yingbin Hua
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao, People’s Republic of China
| | - Chaohui Xie
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao, People’s Republic of China
| | - Haifeng Xu
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao, People’s Republic of China
- Qingdao Eye Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao, People’s Republic of China
| |
Collapse
|
4
|
Shen G, Li Y, Zeng Y, Hong F, Zhang J, Wang Y, Zhang C, Xiang W, Wang J, Fang Z, Qi W, Yang X, Gao G, Zhou T. Kallistatin Deficiency Induces the Oxidative Stress-Related Epithelial-Mesenchymal Transition of Retinal Pigment Epithelial Cells: A Novel Protagonist in Age-Related Macular Degeneration. Invest Ophthalmol Vis Sci 2023; 64:15. [PMID: 37682567 PMCID: PMC10500364 DOI: 10.1167/iovs.64.12.15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 08/15/2023] [Indexed: 09/09/2023] Open
Abstract
Purpose Retinal pigment epithelium (RPE) dysfunction induced by oxidative stress-related epithelial-mesenchymal transition (EMT) of RPE is the primary underlying mechanism of age-related macular degeneration (AMD). Kallistatin (KAL) is a secreted protein with an antioxidative stress effect. However, the relationship between KAL and EMT in RPE has not been determined. Therefore we aimed to explore the impact and mechanism of KAL in oxidative stress-induced EMT of RPE. Methods Sodium iodate (SI) was injected intraperitoneally to construct the AMD rat model and investigate the changes in RPE morphology and KAL expression. KAL knockout rats and KAL transgenic mice were used to explain the effects of KAL on EMT and oxidative stress. In addition, Snail overexpressed adenovirus and si-RNA transfected ARPE19 cells to verify the involvement of Snail in mediating KAL-suppressed EMT of RPE. Results AMD rats induced by SI expressed less KAL in the retina, and KAL knockout rats showed RPE dysfunction spontaneously where EMT and reactive oxygen species (ROS) production increased in RPE. In contrast, KAL overexpression attenuated EMT and ROS levels in RPE, even in TGF-β treatment. Mechanistically, Snail reversed the beneficial effect of KAL on EMT and ROS reduction. Moreover, KAL ameliorated SI-induced AMD-like pathological changes. Conclusions Our findings demonstrated that KAL inhibits oxidative stress-induced EMT by downregulating the transcription factor Snail. Herein, KAL knockout rats may be an appropriate animal model for observing spontaneous RPE dysfunction for AMD-like retinopathy, and KAL may represent a novel therapeutic target for treating dry AMD.
Collapse
Affiliation(s)
- Gang Shen
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
- Department of Laboratory Medicine, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yanmei Li
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Yongcheng Zeng
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Fuyan Hong
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Jing Zhang
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Yan Wang
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Chengwei Zhang
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Wei Xiang
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Jinhong Wang
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Zhenzhen Fang
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Weiwei Qi
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Xia Yang
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Guoquan Gao
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
- Guangdong Engineering & Technology Research Center for Gene Manipulation and Biomacromolecular Products, Sun Yat-Sen University, Guangzhou, China
| | - Ti Zhou
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
- China Key Laboratory of Tropical Disease Control (Sun Yat-Sen University), Ministry of Education, Guangzhou, China
| |
Collapse
|
5
|
Wang Y, Chen Y, Liang J, Jiang M, Zhang T, Wan X, Wu J, Li X, Chen J, Sun J, Hu Y, Huang P, Feng J, Liu T, Sun X. METTL3-mediated m6A modification of HMGA2 mRNA promotes subretinal fibrosis and epithelial-mesenchymal transition. J Mol Cell Biol 2023; 15:mjad005. [PMID: 36945110 PMCID: PMC10603769 DOI: 10.1093/jmcb/mjad005] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 10/01/2022] [Accepted: 11/28/2022] [Indexed: 03/23/2023] Open
Abstract
Subretinal fibrosis is a major cause of the poor visual prognosis for patients with neovascular age-related macular degeneration (nAMD). Myofibroblasts originated from retinal pigment epithelial (RPE) cells through epithelial-mesenchymal transition (EMT) contribute to the fibrosis formation. N6-Methyladenosine (m6A) modification has been implicated in the EMT process and multiple fibrotic diseases. The role of m6A modification in EMT-related subretinal fibrosis has not yet been elucidated. In this study, we found that during subretinal fibrosis in the mouse model of laser-induced choroidal neovascularization, METTL3 was upregulated in RPE cells. Through m6A epitranscriptomic microarray and further verification, high-mobility group AT-hook 2 (HMGA2) was identified as the key downstream target of METTL3, subsequently activating potent EMT-inducing transcription factor SNAIL. Finally, by subretinal injections of adeno-associated virus vectors, we confirmed that METTL3 deficiency in RPE cells could efficiently attenuate subretinal fibrosis in vivo. In conclusion, our present research identified an epigenetic mechanism of METTL3-m6A-HMGA2 in subretinal fibrosis and EMT of RPE cells, providing a novel therapeutic target for subretinal fibrosis secondary to nAMD.
Collapse
Affiliation(s)
- Yuwei Wang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai 200080, China
| | - Yuhong Chen
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai 200080, China
| | - Jian Liang
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai 200080, China
| | - Mei Jiang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai 200080, China
| | - Ting Zhang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai 200080, China
| | - Xiaoling Wan
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai 200080, China
| | - Jiahui Wu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai 200080, China
| | - Xiaomeng Li
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai 200080, China
| | - Jieqiong Chen
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai 200080, China
| | - Junran Sun
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai 200080, China
| | - Yifan Hu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai 200080, China
| | - Peirong Huang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai 200080, China
| | - Jingyang Feng
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai 200080, China
| | - Te Liu
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200031, China
| | - Xiaodong Sun
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai 200080, China
- National Clinical Research Center for Eye Diseases, Shanghai 200080, China
- Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai 200080, China
| |
Collapse
|
6
|
Chien HW, Chen YS, Wang K, Chiou HL, Yang SF, Hsieh YH. Norcantharidin attenuates epidermal growth factor-induced proliferation, EMT and motility in ARPE-19 cells by modulating the AKT/snail/E-cadherin axis. Life Sci 2022; 311:121157. [DOI: 10.1016/j.lfs.2022.121157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 10/27/2022] [Accepted: 11/04/2022] [Indexed: 11/09/2022]
|
7
|
Induction of mesenchymal-epithelial transition (MET) by epigallocatechin-3-gallate to reverse epithelial-mesenchymal transition (EMT) in SNAI1-overexpressed renal cells: A potential anti-fibrotic strategy. J Nutr Biochem 2022; 107:109066. [DOI: 10.1016/j.jnutbio.2022.109066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 03/04/2022] [Accepted: 04/25/2022] [Indexed: 11/19/2022]
|
8
|
Shen G, Li Y, Hong F, Zhang J, Fang Z, Xiang W, Qi W, Yang X, Gao G, Zhou T. A role for Snail-MnSOD axis in regulating epithelial-to-mesenchymal transition markers expression in RPE cells. Biochem Biophys Res Commun 2021; 585:146-154. [PMID: 34808498 DOI: 10.1016/j.bbrc.2021.11.039] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 11/10/2021] [Indexed: 12/16/2022]
Abstract
Age-related macular degeneration (AMD) is a common cause of vision loss. The epithelial-mesenchymal transition (EMT) of retinal pigment epithelial (RPE) cells, accompanied by oxidative damage, plays a crucial role in AMD. It is well known that manganese superoxide dismutase (MnSOD) encoded by SOD2 is a critical molecule in fighting against oxidative stress, and Snail encoded by SNAI1 is the essential transcription factor for EMT. However, the effect of MnSOD on EMT and the underlying mechanism in RPE cells remains unknown. In this study, we found that MnSOD knockdown triggered the EMT by upregulating Snail, while MnSOD overexpression reversed EMT even with TGFβ treatment in RPE cells, and the anti-oxidative stress activity of MnSOD mediated this observation. In addition, Snail depletion increased both expression and activity of MnSOD while Snail overexpression decreased MnSOD expression and activity, and Dual-luciferase reporter and ChIP assays showed that Snail directly bound to E-box (CACCTG) in the SOD2 promoter. Moreover, MnSOD over-expression and Snail interference co-treatment strengthened the anti-oxidation and EMT reversing. Therefore, our findings demonstrate that MnSOD prevents EMT of RPE cells in AMD through inhibiting oxidative injury to RPE. Moreover, a critical EMT transcription factor, Snail, functions as a new negative transcriptional factor of SOD2. Herein, the Snail-MnSOD axis forms a mutual loop in the development of AMD, which may be a novel systemic treatment target for preventing AMD.
Collapse
Affiliation(s)
- Gang Shen
- Program of Molecular Medicine, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China; Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China; Department of Laboratory Medicine, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yanmei Li
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Fuyan Hong
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jing Zhang
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Zhenzhen Fang
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Wei Xiang
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Weiwei Qi
- Program of Molecular Medicine, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China; Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xia Yang
- Program of Molecular Medicine, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China; Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Guoquan Gao
- Program of Molecular Medicine, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China; Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China; Guangdong Engineering & Technology Research Center for Gene Manipulation and Biomacromolecular Products, Sun Yat-sen University, Guangzhou, China
| | - Ti Zhou
- Program of Molecular Medicine, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China; Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China; China Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China.
| |
Collapse
|
9
|
da Silva RA, Roda VMDP, Matsuda M, Siqueira PV, Lustoza-Costa GJ, Wu DC, Hamassaki DE. Cellular components of the idiopathic epiretinal membrane. Graefes Arch Clin Exp Ophthalmol 2021; 260:1435-1444. [PMID: 34842983 DOI: 10.1007/s00417-021-05492-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 10/24/2021] [Accepted: 11/04/2021] [Indexed: 01/19/2023] Open
Abstract
Idiopathic epiretinal membrane (iERM) is a fibrocellular proliferation on the inner surface of the retina, which leads to decreased visual acuity and even central visual loss. As iERM is associated to advanced age and posterior vitreous detachment, a higher prevalence is expected with increasing life expectancy and aging of the global population. Although various cell types of retinal and extra-retinal origin have been described in iERMs (Müller glial cells, astrocytes, hyalocytes, retinal pigment epithelium cells, myofibroblasts, and fibroblasts), myofibroblasts have a central role in collagen production and contractile activity. Thus, myofibroblast differentiation is considered a key event for the iERM formation and progression, and fibroblasts, Müller glial cells, hyalocytes, and retinal pigment epithelium have been identified as myofibroblast precursors. On the other side, the different cell types synthesize growth factors, cytokines, and extracellular matrix, which have a crucial role in ERM pathogenesis. In the present review, the major cellular components and their functions are summarized, and their possible roles in the iERM formation are discussed. By exploring in detail the cellular and molecular aspects of iERM, we seek to contribute for better understanding of this fibrotic disease and the origin of myofibroblasts, which may eventually drive to more targeted therapeutic approaches.
Collapse
Affiliation(s)
- Rafael André da Silva
- Department of Cell & Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, 05508-000, Brazil
| | - Vinicius Moraes de Paiva Roda
- Department of Cell & Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, 05508-000, Brazil
| | - Monique Matsuda
- Laboratory of Investigation in Ophthalmology (LIM-33), Division of Ophthalmology, Faculty of Medicine, University of São Paulo, São Paulo, SP, Brazil
| | - Paula Veloso Siqueira
- Department of Cell & Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, 05508-000, Brazil
| | - Gabriela Jesus Lustoza-Costa
- Department of Cell & Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, 05508-000, Brazil
| | - Davi Chen Wu
- Department of Cell & Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, 05508-000, Brazil.,Department of Ophthalmology, Irmandade de Misericórdia da Santa Casa de São Paulo, São Paulo, SP, Brazil
| | - Dânia Emi Hamassaki
- Department of Cell & Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, 05508-000, Brazil.
| |
Collapse
|
10
|
Park SY, Song WC, Kim B, Oh JW, Park G. Nano-Graphene Oxide-Promoted Epithelial-Mesenchymal Transition of Human Retinal Pigment Epithelial Cells through Regulation of Phospholipase D Signaling. NANOMATERIALS 2021; 11:nano11102546. [PMID: 34684987 PMCID: PMC8540736 DOI: 10.3390/nano11102546] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/16/2021] [Accepted: 09/23/2021] [Indexed: 12/15/2022]
Abstract
Nano-graphene oxide (Nano-GO) is an extensively studied multifunctional carbon nanomaterial with attractive applications in biomedicine and biotechnology. However, few studies have been conducted to assess the epithelial-to-mesenchymal transition (EMT) in the retinal pigment epithelium (RPE). We aimed to determine whether Nano-GO induces EMT by regulating phospholipase D (PLD) signaling in human RPE (ARPE-19) cells. The physicochemical characterization of Nano-GO was performed using a Zetasizer, X-ray diffraction, Fourier-transform infrared spectroscopy, and transmission electron microscopy. RPE cell viability assays were performed, and the migratory effects of RPE cells were evaluated. RPE cell collagen gel contraction was also determined. Intracellular reactive oxygen species (ROS) levels were determined by fluorescence microscopy and flow cytometry. Immunofluorescence staining and western blot analysis were used to detect EMT-related protein expression. Phospholipase D (PLD) enzymatic activities were also measured. Nano-GO significantly enhanced the scratch-healing ability of RPE cells, indicating that the RPE cell migration ability was increased. Following Nano-GO treatment, the RPE cell penetration of the chamber was significantly promoted, suggesting that the migratory ability was strengthened. We also observed collagen gel contraction and the generation of intracellular ROS in RPE cells. The results showed that Nano-GO induced collagen gel contraction and intracellular ROS production in RPE cells. Moreover, immunofluorescence staining and western blot analysis revealed that Nano-GO significantly regulated key molecules of EMT, including epithelial-cadherin, neural-cadherin, α-smooth muscle actin, vimentin, and matrix metalloproteinases (MMP-2 and MMP-9). Interestingly, Nano-GO-induced RPE cell migration and intracellular ROS production were abrogated in PLD-knockdown RPE cells, indicating that PLD activation played a crucial role in the Nano-GO-induced RPE EMT process. We demonstrate for the first time that Nano-GO promotes RPE cell migration through PLD-mediated ROS production. We provide preliminary evidence to support the hypothesis that Nano-GO has adverse health effects related to RPE damage.
Collapse
Affiliation(s)
- Sun Young Park
- Bio-IT Fusion Technology Research Institute, Pusan National University, Busan 46241, Korea;
- Correspondence: (S.Y.P.); (G.P.); Tel.: +82-51-510-3630 (S.Y.P.); +82-51-510-3740 (G.P.); Fax: +82-51-514-7065 (S.Y.P.); +82-51-518-4113 (G.P.)
| | - Woo Chang Song
- Department of Nanofusion Technology, Pusan National University, Busan 46241, Korea; (W.C.S.); (J.-W.O.)
| | - Beomjin Kim
- Bio-IT Fusion Technology Research Institute, Pusan National University, Busan 46241, Korea;
| | - Jin-Woo Oh
- Department of Nanofusion Technology, Pusan National University, Busan 46241, Korea; (W.C.S.); (J.-W.O.)
| | - Geuntae Park
- Department of Nanofusion Technology, Pusan National University, Busan 46241, Korea; (W.C.S.); (J.-W.O.)
- Correspondence: (S.Y.P.); (G.P.); Tel.: +82-51-510-3630 (S.Y.P.); +82-51-510-3740 (G.P.); Fax: +82-51-514-7065 (S.Y.P.); +82-51-518-4113 (G.P.)
| |
Collapse
|
11
|
Snail Upregulates Transcription of FN, LEF, COX2, and COL1A1 in Hepatocellular Carcinoma: A General Model Established for Snail to Transactivate Mesenchymal Genes. Cells 2021; 10:cells10092202. [PMID: 34571852 PMCID: PMC8467536 DOI: 10.3390/cells10092202] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 08/14/2021] [Accepted: 08/22/2021] [Indexed: 12/21/2022] Open
Abstract
SNA is one of the essential EMT transcriptional factors capable of suppressing epithelial maker while upregulating mesenchymal markers. However, the mechanisms for SNA to transactivate mesenchymal markers was not well elucidated. Recently, we demonstrated that SNA collaborates with EGR1 and SP1 to directly upregulate MMP9 and ZEB1. Remarkably, a SNA-binding motif (TCACA) upstream of EGR/SP1 overlapping region on promoters was identified. Herein, we examined whether four other mesenchymal markers, lymphoid enhancer-binding factor (LEF), fibronectin (FN), cyclooxygenase 2 (COX2), and collagen type alpha I (COL1A1) are upregulated by SNA in a similar fashion. Expectedly, SNA is essential for expression of these mesenchymal genes. By deletion mapping and site directed mutagenesis coupled with dual luciferase promoter assay, SNA-binding motif and EGR1/SP1 overlapping region are required for TPA-induced transcription of LEF, FN, COX2 and COL1A1. Consistently, TPA induced binding of SNA and EGR1/SP1 on relevant promoter regions of these mesenchymal genes using ChIP and EMSA. Thus far, we found six of the mesenchymal genes are transcriptionally upregulated by SNA in the same fashion. Moreover, comprehensive screening revealed similar sequence architectures on promoter regions of other SNA-upregulated mesenchymal markers, suggesting that a general model for SNA-upregulated mesenchymal genes can be established.
Collapse
|
12
|
Su Y, Tang Z, Wang F. Role of LINC01592 in TGF-β1-induced epithelial-mesenchymal transition of retinal pigment epithelial cells. Aging (Albany NY) 2021; 13:14053-14064. [PMID: 34032608 PMCID: PMC8202905 DOI: 10.18632/aging.203023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 03/23/2021] [Indexed: 11/25/2022]
Abstract
Regulation of long-chain non-coding RNA01592 (LINC01592) in the process of transforming retinal pigment epithelial (RPE) cells into mesenchymal cells following induction by transforming growth factor beat 1 (TGF-β1) was investigated by interfering with LINC01592 expression in human RPE (hRPE) cells. LINC01592 expression in hRPE cells was significantly increased following treatment with 10 ng/mL TGF-β1 for 48 h. Expression of E-cadherin and Snail were decreased in hRPE cells following induction with TGF-β1 compared with the control group (P < 0.05). Following induction by TGF-β1, expression of E-cadherin, alpha-smooth muscle actin (α-SMA), and Snail were significantly lower in the LINC01592-knockdown group compared with the negative control group (P < 0.05). LINC01592 overexpression significantly enhanced the viability, proliferation, and migration of hRPE cells induced by TGF-β1 (P < 0.05). Following induction by TGF-β1, E-cadherin expression was significantly decreased and α-SMA and Snail expression were significantly increased in the LINC01592-overexpression group compared with the negative control group (P < 0.05). RPE cells induced by TGF-β1 exhibited epithelial-mesenchymal transition (EMT). Inhibiting LINC01592 expression could significantly reduce TGF-β1-induced EMT of hRPE cells. The regulatory effect of LINC01592 on EMT in hRPE cells induced by TGF-β1 provides a novel treatment for proliferative vitreoretinopathy.
Collapse
Affiliation(s)
- Ying Su
- Department of Ophthalmology, The First Hospital of Harbin Medical University, Harbin, China
| | - Ziyan Tang
- Department of Ophthalmology, The First Hospital of Harbin Medical University, Harbin, China
| | - Feng Wang
- Department of Ophthalmology, The First Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
13
|
Zhou Y, Hill C, Yao L, Li J, Hancock D, Downward J, Jones MG, Davies DE, Ewing RM, Skipp P, Wang Y. Quantitative Proteomic Analysis in Alveolar Type II Cells Reveals the Different Capacities of RAS and TGF-β to Induce Epithelial-Mesenchymal Transition. Front Mol Biosci 2021; 8:595712. [PMID: 33869273 PMCID: PMC8048883 DOI: 10.3389/fmolb.2021.595712] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 01/15/2021] [Indexed: 12/12/2022] Open
Abstract
Alveolar type II (ATII) epithelial cells function as stem cells, contributing to alveolar renewal, repair and cancer. Therefore, they are a highly relevant model for studying a number of lung diseases, including acute injury, fibrosis and cancer, in which signals transduced by RAS and transforming growth factor (TGF)-β play critical roles. To identify downstream molecular events following RAS and/or TGF-β activation, we performed proteomic analysis using a quantitative label-free approach (LC-HDMSE) to provide in-depth proteome coverage and estimates of protein concentration in absolute amounts. Data are available via ProteomeXchange with identifier PXD023720. We chose ATIIER:KRASV12 as an experimental cell line in which RAS is activated by adding 4-hydroxytamoxifen (4-OHT). Proteomic analysis of ATII cells treated with 4-OHT or TGF-β demonstrated that RAS activation induces an epithelial–mesenchymal transition (EMT) signature. In contrast, under the same conditions, activation of TGF-β signaling alone only induces a partial EMT. EMT is a dynamic and reversible biological process by which epithelial cells lose their cell polarity and down-regulate cadherin-mediated cell–cell adhesion to gain migratory properties, and is involved in embryonic development, wound healing, fibrosis and cancer metastasis. Thus, these results could help to focus research on the identification of processes that are potentially driving EMT-related human disease.
Collapse
Affiliation(s)
- Yilu Zhou
- Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, United Kingdom.,Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
| | - Charlotte Hill
- Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, United Kingdom
| | - Liudi Yao
- Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, United Kingdom
| | - Juanjuan Li
- Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, United Kingdom
| | - David Hancock
- Oncogene Biology, The Francis Crick Institute, London, United Kingdom
| | - Julian Downward
- Oncogene Biology, The Francis Crick Institute, London, United Kingdom
| | - Mark G Jones
- Institute for Life Sciences, University of Southampton, Southampton, United Kingdom.,Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom.,NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, United Kingdom
| | - Donna E Davies
- Institute for Life Sciences, University of Southampton, Southampton, United Kingdom.,Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom.,NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, United Kingdom
| | - Rob M Ewing
- Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, United Kingdom.,Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
| | - Paul Skipp
- Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, United Kingdom.,Institute for Life Sciences, University of Southampton, Southampton, United Kingdom.,Centre for Proteomic Research, Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
| | - Yihua Wang
- Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, United Kingdom.,Institute for Life Sciences, University of Southampton, Southampton, United Kingdom.,NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, United Kingdom
| |
Collapse
|
14
|
Zhou L, Shi DP, Chu WJ, Yang LL, Xu HF. LRG1 promotes epithelial-mesenchymal transition of retinal pigment epithelium cells by activating NOX4. Int J Ophthalmol 2021; 14:349-355. [PMID: 33747808 DOI: 10.18240/ijo.2021.03.03] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 10/14/2020] [Indexed: 12/19/2022] Open
Abstract
AIM To investigate the effect of leucine-rich-alpha-2-glycoprotein 1 (LRG1) on epithelial-mesenchymal transition (EMT) in retinal pigment epithelium (RPE) cells, and to explore the role of NADPH oxidase 4 (NOX4). METHODS RPE cells (ARPE-19 cell line) were treated with transforming growth factor-β1 (TGF-β1) to induce EMT. Changes of the mRNA and protein expression levels of LRG1 were tested in the TGF-β1 treated cells. The recombinant human LRG1 protein (rLRG1) and siRNA of LRG1 were used to establish accumulation of exogenous LRG1 model and the down-regulation of LRG1 model in ARPE-19 cells respectively, and to detect EMT-related markers including fibronectin, α-smooth muscle actin (α-SMA) and zonula occludens-1 (ZO-1). The mRNA and protein expression level of NOX4 were measured according to the above treatments. VAS2870 was used as a NOX4 inhibitor in rLRG1-treated cells. EMT-related markers were detected to verify the effect of NOX4 in the process of EMT. RESULTS TGF-β1 promoted the expression of LRG1 at both the mRNA and protein levels during the process of EMT which showed the up-regulation of fibronectin and α-SMA, as well as the down-regulation of ZO-1. Furthermore, the rLRG1 promoted EMT of ARPE-19 cells, which manifested high levels of fibronectin and α-SMA and low level of ZO-1, whereas knockdown of LRG1 prevented EMT by decreasing the expressions of fibronectin and α-SMA and increasing the expression of ZO-1 in ARPE-19 cells. Besides, the rLRG1 activated and LRG1 siRNA suppressed NOX4 expression. EMT was inhibited when VAS2870 was used in the rLRG1-treated cells. CONCLUSION These results for the first time demonstrate that LRG1 promotes EMT of RPE cells by activating NOX4, which may provide a novel direction to explore the mechanisms of subretinal fibrosis.
Collapse
Affiliation(s)
- Li Zhou
- Medical College, Qingdao University, Qingdao 266071, Shandong Province, China.,State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao 266071, Shandong Province, China
| | - De-Peng Shi
- Qingdao Eye Hospital, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao 266071, Shandong Province, China
| | - Wen-Juan Chu
- Qingdao Eye Hospital, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao 266071, Shandong Province, China
| | - Ling-Ling Yang
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao 266071, Shandong Province, China
| | - Hai-Feng Xu
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao 266071, Shandong Province, China.,Qingdao Eye Hospital, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao 266071, Shandong Province, China
| |
Collapse
|
15
|
Epithelial-Mesenchymal Transition and Senescence in the Retinal Pigment Epithelium of NFE2L2/PGC-1α Double Knock-Out Mice. Int J Mol Sci 2021; 22:ijms22041684. [PMID: 33567500 PMCID: PMC7915526 DOI: 10.3390/ijms22041684] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 02/04/2021] [Indexed: 12/13/2022] Open
Abstract
Age-related macular degeneration (AMD) is the most prevalent form of irreversible blindness worldwide in the elderly population. In our previous studies, we found that deficiencies in the nuclear factor, erythroid 2 like 2 (NFE2L2) and peroxisome proliferator-activated receptor gamma coactivator 1-α (PGC-1α) genes caused AMD-like pathological phenotypes in mice. In the present work, we show hijacked epithelial-mesenchymal transition (EMT) due to the common loss of PGC-1α and NFE2L2 (double knock-out, dKO) genes in aged animals. The implanted area was assessed by histology, immunohistochemistry and transmission electron microscopy. Confocal microscopy revealed altered regions in the filamentous actin ring. This contrasted with hexagonal RPE morphology in wild-type mice. The ultrastructural RPE features here illustrated loss of apical microvilli, alteration of cell-cell contact, loss of basal in-folding with deposits on Bruch’s membrane, and excessive lipofuscin deposition in dKO samples. We also found the expression of epithelial-mesenchymal transition transcription factors, such as Snail, Slug, collagen 1, vimentin and OB-cadherin, to be significantly different in dKO RPEs. An increased immunoreactivity of senescence markers p16, DEC1 and HMGB1 was also noted. These findings suggest that EMT and senescence pathways may intersect in the retinas of dKO mice. Both processes can be activated by damage to the RPE, which may be caused by increased oxidative stress resulting from the absence of NFE2L2 and PGC-1α genes, important for antioxidant defense. This dKO model may provide useful tools for studying AMD pathogenesis and evaluating novel therapies for this disease.
Collapse
|
16
|
Zou H, Shan C, Ma L, Liu J, Yang N, Zhao J. Polarity and epithelial-mesenchymal transition of retinal pigment epithelial cells in proliferative vitreoretinopathy. PeerJ 2020; 8:e10136. [PMID: 33150072 PMCID: PMC7583629 DOI: 10.7717/peerj.10136] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 09/18/2020] [Indexed: 12/11/2022] Open
Abstract
Under physiological conditions, retinal pigment epithelium (RPE) is a cellular monolayer composed of mitotically quiescent cells. Tight junctions and adherens junctions maintain the polarity of RPE cells, and are required for cellular functions. In proliferative vitreoretinopathy (PVR), upon retinal tear, RPE cells lose cell-cell contact, undergo epithelial-mesenchymal transition (EMT), and ultimately transform into myofibroblasts, leading to the formation of fibrocellular membranes on both surfaces of the detached retina and on the posterior hyaloids, which causes tractional retinal detachment. In PVR, RPE cells are crucial contributors, and multiple signaling pathways, including the SMAD-dependent pathway, Rho pathway, MAPK pathways, Jagged/Notch pathway, and the Wnt/β-catenin pathway are activated. These pathways mediate the EMT of RPE cells, which play a key role in the pathogenesis of PVR. This review summarizes the current body of knowledge on the polarized phenotype of RPE, the role of cell-cell contact, and the molecular mechanisms underlying the RPE EMT in PVR, emphasizing key insights into potential approaches to prevent PVR.
Collapse
Affiliation(s)
- Hui Zou
- Eye Center, The Second Hospital of Jilin University, Changchun, China
| | - Chenli Shan
- Eye Center, The Second Hospital of Jilin University, Changchun, China
| | - Linlin Ma
- Eye Center, The Second Hospital of Jilin University, Changchun, China
| | - Jia Liu
- Eye Center, The Second Hospital of Jilin University, Changchun, China
| | - Ning Yang
- Eye Center, The Second Hospital of Jilin University, Changchun, China
| | - Jinsong Zhao
- Eye Center, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
17
|
Zhang Y, Wang K, Pan J, Yang S, Yao H, Li M, Li H, Lei H, Jin H, Wang F. Exosomes mediate an epithelial-mesenchymal transition cascade in retinal pigment epithelial cells: Implications for proliferative vitreoretinopathy. J Cell Mol Med 2020; 24:13324-13335. [PMID: 33047885 PMCID: PMC7701536 DOI: 10.1111/jcmm.15951] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 05/16/2020] [Accepted: 09/16/2020] [Indexed: 12/13/2022] Open
Abstract
Exosomes have recently emerged as a pivotal mediator of many physiological and pathological processes. However, the role of exosomes in proliferative vitreoretinopathy (PVR) has not been reported. In this study, we aimed to investigate the role of exosomes in PVR. Transforming growth factor beta 2 (TGFß-2) was used to induce epithelial-mesenchymal transition (EMT) of retinal pigment epithelial (RPE) cells, as an in vitro model of PVR. Exosomes from normal and EMTed RPE cells were extracted and identified. We incubated extracted exosomes with recipient RPE cells, and co-cultured EMTed RPE cells and recipient RPE cells in the presence of the exosome inhibitor GW4869. Both experiments suggested that there are further EMT-promoting effects of exosomes from EMTed RPE cells. MicroRNA sequencing was also performed to identify the miRNA profiles in exosomes from both groups. We identified 34 differentially expressed exosomal miRNAs (P <. 05). Importantly, miR-543 was found in exosomes from EMTed RPE cells, and miR-543-enriched exosomes significantly induced the EMT of recipient RPE cells. Our study demonstrates that exosomal miRNA is differentially expressed in RPE cells during EMT and that these exosomal miRNAs may play pivotal roles in EMT induction. Our results highlight the importance of exosomes as cellular communicators within the microenvironment of PVR.
Collapse
Affiliation(s)
- Yao Zhang
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Kaizhe Wang
- Division of Physical Biology and Bioimaging Center, Shanghai Synchrotron Radiation Facility, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai, China
| | - Jiabin Pan
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shuai Yang
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Haipei Yao
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Min Li
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hui Li
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hetian Lei
- Department of Ophthalmology, Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Haiying Jin
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Fang Wang
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
18
|
Ilimaquinone inhibits neovascular age-related macular degeneration through modulation of Wnt/β-catenin and p53 pathways. Pharmacol Res 2020; 161:105146. [PMID: 32814173 DOI: 10.1016/j.phrs.2020.105146] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 08/11/2020] [Accepted: 08/11/2020] [Indexed: 11/22/2022]
Abstract
Neovascular age-related macular degeneration (nAMD) is a common cause of irreversible vision loss in the elderly. Anti-vascular endothelial growth factor has been effective in treating pathological ocular neovascularization, but it has limitations including the need for repeated intraocular injections for the maintenance of therapeutic effects in most patients and poor or non-response to this agent in some patients. in vitro cellular studies were conducted using retinal pigment epithelial cell lines (ARPE-19 and hTERT-RPE1), human umbilical vein endothelial cells (HUVECs), and human umbilical vein smooth muscle cells (HUVSMCs). in vivo efficacy of ilimaquinone (IQ) was tested in laser-induced choroidal neovascularization mouse and rabbit models. Tissue distribution study was performed in male C57BL6/J mice. IQ, 4,9-friedodrimane-type sesquiterpenoid isolated from the marine sponge, repressed the expression of angiogenic/inflammatory factors and restored the expression of E-cadherin in retinal pigment epithelial cells by inhibiting the Wnt/β-catenin pathway. In addition, it selectively inhibited proliferation and tube formation of HUVECs by activating the p53 pathway. Topical and intraperitoneal administration of IQ significantly reduced choroidal neovascularization in rabbits and mice with laser-induced choroidal neovascularization. Notably, IQ by the oral route of exposure was highly permeable to the eyes and suppressed abnormal vascular leakage by downregulation of β-catenin and stabilization of p53 in vivo. Our findings demonstrate that IQ functions through regulation of p53 and Wnt/β-catenin pathways with conceivable advantages over existing cytokine-targeted anti-angiogenic therapies.
Collapse
|
19
|
Intracellular amyloid-β disrupts tight junctions of the retinal pigment epithelium via NF-κB activation. Neurobiol Aging 2020; 95:115-122. [PMID: 32795848 DOI: 10.1016/j.neurobiolaging.2020.07.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 07/07/2020] [Accepted: 07/14/2020] [Indexed: 12/16/2022]
Abstract
Drusen are focal deposits between the retinal pigment epithelium (RPE) and Bruch's membrane in the retina of patients with age-related macular degeneration. Amyloid-β is one of the important components of drusen, which leads to local inflammation. Furthermore, intracellular amyloid-β disrupts tight junctions of the RPE. However, the intracellular mechanisms linking intracellular amyloid-β and tight-junction disruption are not clear. In this study, intracellular amyloid-β oligomers activated nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) p65, leading to the disorganization of tight junctions of the RPE in mice after subretinal injection of amyloid-β. Amyloid-β also triggered NF-κB activation in the RPE cells in confluent culture, which was inhibited by the suppression of the advanced glycosylation end product-specific receptor. NF-κB inhibition by an IκB kinase inhibitor prevented the suppression of expression of tight-junction proteins, zonula occuludens-1 and occludin in RPE cells. In addition, tight-junction complexes remained intact in the RPE of mice with NF-κB inhibition, although there were intracellular amyloid-β oligomers. These data suggested that NF-κB inhibition might be a therapeutic approach to prevent amyloid-β-mediated tight-junction disruption.
Collapse
|
20
|
Micati DJ, Radhakrishnan K, Young JC, Rajpert‐De Meyts E, Hime GR, Abud HE, Loveland KL. ‘Snail factors in testicular germ cell tumours and their regulation by the BMP4 signalling pathway’. Andrology 2020; 8:1456-1470. [DOI: 10.1111/andr.12823] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 04/20/2020] [Accepted: 05/14/2020] [Indexed: 12/19/2022]
Affiliation(s)
- Diana J. Micati
- Centre for Reproductive Health Hudson Institute of Medical Research Clayton Victoria Australia
- Department of Molecular and Translational Sciences Monash University Clayton Victoria Australia
| | - Karthika Radhakrishnan
- Centre for Reproductive Health Hudson Institute of Medical Research Clayton Victoria Australia
- Department of Molecular and Translational Sciences Monash University Clayton Victoria Australia
| | - Julia C. Young
- Centre for Reproductive Health Hudson Institute of Medical Research Clayton Victoria Australia
- Department of Molecular and Translational Sciences Monash University Clayton Victoria Australia
- Department of Anatomy and Developmental Biology Monash Biomedicine Discovery Institute Monash University Clayton Victoria Australia
| | - Ewa Rajpert‐De Meyts
- Department of Growth and Reproduction, Rigshospitalet University of Copenhagen Copenhagen Denmark
| | - Gary R. Hime
- Department of Anatomy and Neuroscience University of Melbourne Melbourne Victoria Australia
| | - Helen E. Abud
- Department of Anatomy and Developmental Biology Monash Biomedicine Discovery Institute Monash University Clayton Victoria Australia
- Stem Cells and Development Program Monash Biomedicine Discovery Institute Monash University Clayton Victoria Australia
| | - Kate L. Loveland
- Centre for Reproductive Health Hudson Institute of Medical Research Clayton Victoria Australia
- Department of Molecular and Translational Sciences Monash University Clayton Victoria Australia
- Department of Anatomy and Developmental Biology Monash Biomedicine Discovery Institute Monash University Clayton Victoria Australia
| |
Collapse
|
21
|
Bao H, Yang S, Li H, Yao H, Zhang Y, Zhang J, Xu G, Jin H, Wang F. The Interplay Between E-Cadherin, Connexin 43, and Zona Occludens 1 in Retinal Pigment Epithelial Cells. Invest Ophthalmol Vis Sci 2020; 60:5104-5111. [PMID: 31826237 DOI: 10.1167/iovs.19-27768] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose Cell-cell contact in retinal pigment epithelium (RPE) involves adherent junctions, gap junctions, and tight junctions, which are primarily composed by E-cadherin, zona occludens 1 (ZO-1), and connexin 43, respectively. Here, we aimed to explore the relationship and interplay between these junction-associated proteins. Methods E-cadherin, connexin 43, and ZO-1 expression in human primary RPE in the early phase after TGF-β1 stimulation was detected. The knockdown of E-cadherin, ZO-1, and connexin 43 was performed to characterize the regulatory network involving these three proteins. Dye transfer and FITC-dextran permeability assays were conducted to observe the epithelial functional alterations. Transmission electron microscopy (TEM) was used to observe the ultrastructure of the cell-cell junctions in mouse RPE. The immunofluorescence staining and coimmunoprecipitation were performed to observe the colocalization and the physical association of E-cadherin, ZO-1, and connexin 43. Results Among these three components, E-cadherin appeared to be the first protein that was downregulated after TGF-β1 treatment. The ultrastructures of adherent junctions, gap junctions, and tight junctions could be observed in mouse RPE by TEM. E-cadherin, ZO-1, and connexin 43 were colocalized and physically bound to each other. The knockdown of one of these three proteins led to downregulation of the other two proteins and compromised epithelial function. Conclusions E-cadherin, ZO-1, and connexin 43 were physically associated with each other and were mutually regulated. To enhance the understanding of cell-cell contacts, a holistic view is needed. Our results provide new insights in RPE disorders such as proliferative vitreoretinopathy.
Collapse
Affiliation(s)
- Huiqian Bao
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Tongji Eye Institute, Tongji University School of Medicine, Shanghai, China.,School of Medicine, Nanchang University, Nanchang, Jiangxi Province, China
| | - Shuai Yang
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Tongji Eye Institute, Tongji University School of Medicine, Shanghai, China
| | - Hui Li
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Tongji Eye Institute, Tongji University School of Medicine, Shanghai, China
| | - Haipei Yao
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Tongji Eye Institute, Tongji University School of Medicine, Shanghai, China
| | - Yao Zhang
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Tongji Eye Institute, Tongji University School of Medicine, Shanghai, China
| | - Jingfa Zhang
- Department of Ophthalmology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Guotong Xu
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Tongji Eye Institute, Tongji University School of Medicine, Shanghai, China
| | - Haiying Jin
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Tongji Eye Institute, Tongji University School of Medicine, Shanghai, China
| | - Fang Wang
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Tongji Eye Institute, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
22
|
Leng Z, Li Y, Zhou G, Lv X, Ai W, Li J, Hou L. Krüppel-like factor 4 regulates stemness and mesenchymal properties of colorectal cancer stem cells through the TGF-β1/Smad/snail pathway. J Cell Mol Med 2020; 24:1866-1877. [PMID: 31830379 PMCID: PMC6991673 DOI: 10.1111/jcmm.14882] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 09/09/2019] [Accepted: 09/18/2019] [Indexed: 12/12/2022] Open
Abstract
Krüppel-like factor 4 (KLF4) was closely associated with epithelial-mesenchymal transition and stemness in colorectal cancer stem cells (CSCs)-enriched spheroid cells. Nonetheless, the underlying molecular mechanism is unclear. This study showed that KLF4 overexpression was accompanied with stemness and mesenchymal features in Lgr5+ CD44+ EpCAM+ colorectal CSCs. KLF4 knockdown suppressed stemness, mesenchymal features and activation of the TGF-β1 pathway, whereas enforced KLF4 overexpression activated TGF-β1, phosphorylation of Smad 2/3 and Snail expression, and restored stemness and mesenchymal phenotypes. Furthermore, TGF-β1 pathway inhibition invalidated KLF4-facilitated stemness and mesenchymal features without affecting KLF4 expression. The data from the current study are the first to demonstrate that KLF4 maintains stemness and mesenchymal properties through the TGF-β1/Smad/Snail pathway in Lgr5+ CD44+ EpCAM+ colorectal CSCs.
Collapse
Affiliation(s)
- Zhengwei Leng
- Northeast Sichuan Acute Pancreatic Research CenterNorth Sichuan Medical CollegeSichuanChina
- Cancer Stem Cells Research CenterAffiliated Hospital of North Sichuan Medical CollegeSichuanChina
| | - Yong Li
- Northeast Sichuan Acute Pancreatic Research CenterNorth Sichuan Medical CollegeSichuanChina
| | - Guojun Zhou
- Northeast Sichuan Acute Pancreatic Research CenterNorth Sichuan Medical CollegeSichuanChina
| | - Xiaojiang Lv
- Northeast Sichuan Acute Pancreatic Research CenterNorth Sichuan Medical CollegeSichuanChina
| | - Walden Ai
- Department of Biology, Chemistry and Environmental Health ScienceBenedict CollegeColumbiaSCUSA
| | - Jianshui Li
- Northeast Sichuan Acute Pancreatic Research CenterNorth Sichuan Medical CollegeSichuanChina
| | - Lingmi Hou
- Northeast Sichuan Acute Pancreatic Research CenterNorth Sichuan Medical CollegeSichuanChina
- Cancer Stem Cells Research CenterAffiliated Hospital of North Sichuan Medical CollegeSichuanChina
- Thyriod and Breast SurgeryAffiliated Hospital of North Sichuan Medical CollegeSichuanChina
| |
Collapse
|
23
|
Liu B, Xing X, Li X, Guo Q, Xu T, Xu K. ZNF259 promotes breast cancer cells invasion and migration via ERK/GSK3β/snail signaling. Cancer Manag Res 2018; 10:3159-3168. [PMID: 30214308 PMCID: PMC6126507 DOI: 10.2147/cmar.s174745] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Purpose Zinc finger protein 259 (ZNF259), also known as ZPR1, is a zinc finger-containing protein that can bind the intracellular tyrosine kinase domain of EGFR. At present, our knowledge on ZNF259 in cancers is limited. Here, we aimed to explore the biological functions of ZNF259 in breast cancer and reveal their mechanisms. Patients and methods The expression of ZNF259 was measured in 133 cases of breast cancer by immunohistochemistry. The online database Kaplan–Meier (KM) Plotter Online Tool was used to analyze the relationship between ZNF259 expression and breast cancer patient survival prognosis. Plasmid transfection and small interfering RNA and inhibitor treatments were carried out to explore the functions of ZNF259 in breast cancer cell lines and its potential mechanism. Matrigel invasion and wound healing assays were performed to detect the invasion and migration ability of cancer cells. In addition, protein expressions in tissues and cells were determined by Western blotting. Results ZNF259 expression was much higher in breast cancer cells than in the adjacent normal breast duct glandular epithelial cells (75.94% vs 7.52%, P<0.001) and was closely related to the breast cancer patients’ TNM stages (P=0.013) and lymph node metastasis (P=0.021). Knockdown of ZNF259 could downregulate p-ERK, p-GSK3β, and Snail expression, and upregulate the expression of E-cadherin and ZO-1, and then it also inhibited invasion and migration by the breast cancer cell lines MCF-7 and MDA-MB-231. Correspondingly, ZNF259 transfection could upregulate p-ERK, p-GSK3β, and Snail expression, and downregulate E-cadherin and ZO-1 expression, which led to stronger invasion and migration abilities of cancer cells. Furthermore, the ERK inhibitor U0126 could reverse all these effects induced by ZNF259 transfection. Conclusion ZNF259 could promote breast cancer cell invasion and migration by activating the ERK/GSK3β/Snail signaling pathway.
Collapse
Affiliation(s)
- Bin Liu
- Medical Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning Province, People's Republic of China
| | - Xiaojing Xing
- Medical Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning Province, People's Republic of China
| | - Xiang Li
- Breast Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning Province, People's Republic of China
| | - Qianxue Guo
- Medical Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning Province, People's Republic of China
| | - Tonghong Xu
- Medical Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning Province, People's Republic of China
| | - Ke Xu
- Thoracic Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning Province, People's Republic of China,
| |
Collapse
|
24
|
Functional Role of Non-Coding RNAs during Epithelial-To-Mesenchymal Transition. Noncoding RNA 2018; 4:ncrna4020014. [PMID: 29843425 PMCID: PMC6027143 DOI: 10.3390/ncrna4020014] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 05/22/2018] [Accepted: 05/23/2018] [Indexed: 01/17/2023] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) is a key biological process involved in a multitude of developmental and pathological events. It is characterized by the progressive loss of cell-to-cell contacts and actin cytoskeletal rearrangements, leading to filopodia formation and the progressive up-regulation of a mesenchymal gene expression pattern enabling cell migration. Epithelial-to-mesenchymal transition is already observed in early embryonic stages such as gastrulation, when the epiblast undergoes an EMT process and therefore leads to the formation of the third embryonic layer, the mesoderm. Epithelial-to-mesenchymal transition is pivotal in multiple embryonic processes, such as for example during cardiovascular system development, as valve primordia are formed and the cardiac jelly is progressively invaded by endocardium-derived mesenchyme or as the external cardiac cell layer is established, i.e., the epicardium and cells detached migrate into the embryonic myocardial to form the cardiac fibrous skeleton and the coronary vasculature. Strikingly, the most important biological event in which EMT is pivotal is cancer development and metastasis. Over the last years, understanding of the transcriptional regulatory networks involved in EMT has greatly advanced. Several transcriptional factors such as Snail, Slug, Twist, Zeb1 and Zeb2 have been reported to play fundamental roles in EMT, leading in most cases to transcriptional repression of cell⁻cell interacting proteins such as ZO-1 and cadherins and activation of cytoskeletal markers such as vimentin. In recent years, a fundamental role for non-coding RNAs, particularly microRNAs and more recently long non-coding RNAs, has been identified in normal tissue development and homeostasis as well as in several oncogenic processes. In this study, we will provide a state-of-the-art review of the functional roles of non-coding RNAs, particularly microRNAs, in epithelial-to-mesenchymal transition in both developmental and pathological EMT.
Collapse
|
25
|
Pieterse E, Rother N, Garsen M, Hofstra JM, Satchell SC, Hoffmann M, Loeven MA, Knaapen HK, van der Heijden OWH, Berden JHM, Hilbrands LB, van der Vlag J. Neutrophil Extracellular Traps Drive Endothelial-to-Mesenchymal Transition. Arterioscler Thromb Vasc Biol 2017; 37:1371-1379. [PMID: 28495931 DOI: 10.1161/atvbaha.117.309002] [Citation(s) in RCA: 190] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 05/01/2017] [Indexed: 12/12/2022]
Abstract
OBJECTIVE An excessive release and impaired degradation of neutrophil extracellular traps (NETs) leads to the continuous exposure of NETs to the endothelium in a variety of hematologic and autoimmune disorders, including lupus nephritis. This study aims to unravel the mechanisms through which NETs jeopardize vascular integrity. APPROACH AND RESULTS Microvascular and macrovascular endothelial cells were exposed to NETs, and subsequent effects on endothelial integrity and function were determined in vitro and in vivo. We found that endothelial cells have a limited capacity to internalize NETs via the receptor for advanced glycation endproducts. An overflow of the phagocytic capacity of endothelial cells for NETs resulted in the persistent extracellular presence of NETs, which rapidly altered endothelial cell-cell contacts and induced vascular leakage and transendothelial albumin passage through elastase-mediated proteolysis of the intercellular junction protein VE-cadherin. Furthermore, NET-associated elastase promoted the nuclear translocation of junctional β-catenin and induced endothelial-to-mesenchymal transition in cultured endothelial cells. In vivo, NETs could be identified in kidney samples of diseased MRL/lpr mice and patients with lupus nephritis, in whom the glomerular presence of NETs correlated with the severity of proteinuria and with glomerular endothelial-to-mesenchymal transition. CONCLUSIONS These results indicate that an excess of NETs exceeds the phagocytic capacity of endothelial cells for NETs and promotes vascular leakage and endothelial-to-mesenchymal transition through the degradation of VE-cadherin and the subsequent activation of β-catenin signaling. Our data designate NET-associated elastase as a potential therapeutic target in the prevention of endothelial alterations in diseases characterized by aberrant NET release.
Collapse
Affiliation(s)
- Elmar Pieterse
- From the Nephrology Research Laboratory, Department of Nephrology (E.P., N.R., M.G., J.M.H., M.A.L., J.H.M.B., L.B.H., J.v.d.V.), Department of Rheumatology (H.K.K.), and Department of Obstetrics & Gynecology (O.W.H.v.d.H.), Radboud University Medical Center, Nijmegen, The Netherlands; Academic Renal Unit, School of Clinical Science, University of Bristol, United Kingdom (S.C.S.); and Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich Alexander University of Erlangen-Nuremberg, Germany (M.H.)
| | - Nils Rother
- From the Nephrology Research Laboratory, Department of Nephrology (E.P., N.R., M.G., J.M.H., M.A.L., J.H.M.B., L.B.H., J.v.d.V.), Department of Rheumatology (H.K.K.), and Department of Obstetrics & Gynecology (O.W.H.v.d.H.), Radboud University Medical Center, Nijmegen, The Netherlands; Academic Renal Unit, School of Clinical Science, University of Bristol, United Kingdom (S.C.S.); and Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich Alexander University of Erlangen-Nuremberg, Germany (M.H.)
| | - Marjolein Garsen
- From the Nephrology Research Laboratory, Department of Nephrology (E.P., N.R., M.G., J.M.H., M.A.L., J.H.M.B., L.B.H., J.v.d.V.), Department of Rheumatology (H.K.K.), and Department of Obstetrics & Gynecology (O.W.H.v.d.H.), Radboud University Medical Center, Nijmegen, The Netherlands; Academic Renal Unit, School of Clinical Science, University of Bristol, United Kingdom (S.C.S.); and Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich Alexander University of Erlangen-Nuremberg, Germany (M.H.)
| | - Julia M Hofstra
- From the Nephrology Research Laboratory, Department of Nephrology (E.P., N.R., M.G., J.M.H., M.A.L., J.H.M.B., L.B.H., J.v.d.V.), Department of Rheumatology (H.K.K.), and Department of Obstetrics & Gynecology (O.W.H.v.d.H.), Radboud University Medical Center, Nijmegen, The Netherlands; Academic Renal Unit, School of Clinical Science, University of Bristol, United Kingdom (S.C.S.); and Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich Alexander University of Erlangen-Nuremberg, Germany (M.H.)
| | - Simon C Satchell
- From the Nephrology Research Laboratory, Department of Nephrology (E.P., N.R., M.G., J.M.H., M.A.L., J.H.M.B., L.B.H., J.v.d.V.), Department of Rheumatology (H.K.K.), and Department of Obstetrics & Gynecology (O.W.H.v.d.H.), Radboud University Medical Center, Nijmegen, The Netherlands; Academic Renal Unit, School of Clinical Science, University of Bristol, United Kingdom (S.C.S.); and Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich Alexander University of Erlangen-Nuremberg, Germany (M.H.)
| | - Markus Hoffmann
- From the Nephrology Research Laboratory, Department of Nephrology (E.P., N.R., M.G., J.M.H., M.A.L., J.H.M.B., L.B.H., J.v.d.V.), Department of Rheumatology (H.K.K.), and Department of Obstetrics & Gynecology (O.W.H.v.d.H.), Radboud University Medical Center, Nijmegen, The Netherlands; Academic Renal Unit, School of Clinical Science, University of Bristol, United Kingdom (S.C.S.); and Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich Alexander University of Erlangen-Nuremberg, Germany (M.H.)
| | - Markus A Loeven
- From the Nephrology Research Laboratory, Department of Nephrology (E.P., N.R., M.G., J.M.H., M.A.L., J.H.M.B., L.B.H., J.v.d.V.), Department of Rheumatology (H.K.K.), and Department of Obstetrics & Gynecology (O.W.H.v.d.H.), Radboud University Medical Center, Nijmegen, The Netherlands; Academic Renal Unit, School of Clinical Science, University of Bristol, United Kingdom (S.C.S.); and Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich Alexander University of Erlangen-Nuremberg, Germany (M.H.)
| | - Hanneke K Knaapen
- From the Nephrology Research Laboratory, Department of Nephrology (E.P., N.R., M.G., J.M.H., M.A.L., J.H.M.B., L.B.H., J.v.d.V.), Department of Rheumatology (H.K.K.), and Department of Obstetrics & Gynecology (O.W.H.v.d.H.), Radboud University Medical Center, Nijmegen, The Netherlands; Academic Renal Unit, School of Clinical Science, University of Bristol, United Kingdom (S.C.S.); and Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich Alexander University of Erlangen-Nuremberg, Germany (M.H.)
| | - Olivier W H van der Heijden
- From the Nephrology Research Laboratory, Department of Nephrology (E.P., N.R., M.G., J.M.H., M.A.L., J.H.M.B., L.B.H., J.v.d.V.), Department of Rheumatology (H.K.K.), and Department of Obstetrics & Gynecology (O.W.H.v.d.H.), Radboud University Medical Center, Nijmegen, The Netherlands; Academic Renal Unit, School of Clinical Science, University of Bristol, United Kingdom (S.C.S.); and Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich Alexander University of Erlangen-Nuremberg, Germany (M.H.)
| | - Jo H M Berden
- From the Nephrology Research Laboratory, Department of Nephrology (E.P., N.R., M.G., J.M.H., M.A.L., J.H.M.B., L.B.H., J.v.d.V.), Department of Rheumatology (H.K.K.), and Department of Obstetrics & Gynecology (O.W.H.v.d.H.), Radboud University Medical Center, Nijmegen, The Netherlands; Academic Renal Unit, School of Clinical Science, University of Bristol, United Kingdom (S.C.S.); and Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich Alexander University of Erlangen-Nuremberg, Germany (M.H.)
| | - Luuk B Hilbrands
- From the Nephrology Research Laboratory, Department of Nephrology (E.P., N.R., M.G., J.M.H., M.A.L., J.H.M.B., L.B.H., J.v.d.V.), Department of Rheumatology (H.K.K.), and Department of Obstetrics & Gynecology (O.W.H.v.d.H.), Radboud University Medical Center, Nijmegen, The Netherlands; Academic Renal Unit, School of Clinical Science, University of Bristol, United Kingdom (S.C.S.); and Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich Alexander University of Erlangen-Nuremberg, Germany (M.H.)
| | - Johan van der Vlag
- From the Nephrology Research Laboratory, Department of Nephrology (E.P., N.R., M.G., J.M.H., M.A.L., J.H.M.B., L.B.H., J.v.d.V.), Department of Rheumatology (H.K.K.), and Department of Obstetrics & Gynecology (O.W.H.v.d.H.), Radboud University Medical Center, Nijmegen, The Netherlands; Academic Renal Unit, School of Clinical Science, University of Bristol, United Kingdom (S.C.S.); and Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich Alexander University of Erlangen-Nuremberg, Germany (M.H.).
| |
Collapse
|
26
|
Kim D, Ko HS, Park GB, Hur DY, Kim YS, Yang JW. Vandetanib and ADAM inhibitors synergistically attenuate the pathological migration of EBV-infected retinal pigment epithelial cells by regulating the VEGF-mediated MAPK pathway. Exp Ther Med 2017; 13:1415-1425. [PMID: 28413487 PMCID: PMC5377331 DOI: 10.3892/etm.2017.4110] [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: 10/20/2015] [Accepted: 11/25/2016] [Indexed: 12/11/2022] Open
Abstract
The extracellular signals induced by vascular endothelial growth factor (VEGF) are implicated in choroidal neovascularization (CNV) and thus, are associated with vision-limiting complications in the human retina. Vandetanib is an oral anticancer drug that selectively inhibits the activities of VEGF receptor and epidermal growth factor receptor tyrosine kinase; however, the effects of vandetanib on VEGF in retinal pigment epithelial (RPE) cells have not yet been studied. In the present study, a combined treatment of vandetanib and a disintegrin and metalloproteinase (ADAM) protein inhibitors were used to assess the regulation of Epstein-Barr virus (EBV)-infected ARPE19 cells (ARPE19/EBV) migration as a model of CNV. Vandetanib suppressed the expression of the mesenchymal markers ADAM10 and ADAM17 in ARPE19/EBV cells, and also upregulated epithelial cell markers of the RPE cells, E-cadherin and N-cadherin. The migratory activity of ARPE19/EBV induced by VEGF was efficiently blocked by vandetanib. Furthermore, co-treatment with vandetanib and an ADAM10 inhibitor (GI254023X) or ADAM17 inhibitor (Marimastat) synergistically prevented migration and the expression of vimentin, Snail and α-smooth muscle actin by regulating extracellular signal-regulated kinase and p38 mitogen-activated protein kinase. These results suggest that a combination treatment of vandetanib and ADAM inhibitors may be developed as a novel therapeutic regimen to control retina neovascular disease.
Collapse
Affiliation(s)
- Daejin Kim
- Department of Anatomy, Inje University College of Medicine, Inje University Busan Paik Hospital, Busan 614-735, Republic of Korea.,Ocular Neovascular Disease Research Center, Inje University College of Medicine, Inje University Busan Paik Hospital, Busan 614-735, Republic of Korea
| | - Hyun-Suk Ko
- Department of Anatomy, Inje University College of Medicine, Inje University Busan Paik Hospital, Busan 614-735, Republic of Korea
| | - Ga Bin Park
- Department of Anatomy, Inje University College of Medicine, Inje University Busan Paik Hospital, Busan 614-735, Republic of Korea.,Ocular Neovascular Disease Research Center, Inje University College of Medicine, Inje University Busan Paik Hospital, Busan 614-735, Republic of Korea
| | - Dae Young Hur
- Department of Anatomy, Inje University College of Medicine, Inje University Busan Paik Hospital, Busan 614-735, Republic of Korea.,Ocular Neovascular Disease Research Center, Inje University College of Medicine, Inje University Busan Paik Hospital, Busan 614-735, Republic of Korea
| | - Yeong Seok Kim
- Department of Anatomy, Inje University College of Medicine, Inje University Busan Paik Hospital, Busan 614-735, Republic of Korea
| | - Jae Wook Yang
- Department of Ophthalmology, Inje University College of Medicine, Inje University Busan Paik Hospital, Busan 614-735, Republic of Korea.,Ocular Neovascular Disease Research Center, Inje University College of Medicine, Inje University Busan Paik Hospital, Busan 614-735, Republic of Korea
| |
Collapse
|
27
|
Kim JH, Park S, Chung H, Oh S. Wnt5a attenuates the pathogenic effects of the Wnt/β-catenin pathway in human retinal pigment epithelial cells via down-regulating β-catenin and Snail. BMB Rep 2016; 48:525-30. [PMID: 26246285 PMCID: PMC4641237 DOI: 10.5483/bmbrep.2015.48.9.140] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Indexed: 12/19/2022] Open
Abstract
Activation of the Wnt/β-catenin pathway plays a pathogenic role in age-related macular degeneration (AMD) and is thus a potential target for the development of therapeutics for this disease. Here, we demonstrated that Wnt5a antagonized β-catenin response transcription (CRT) induced with Wnt3a by promoting β-catenin phosphorylation at Ser33/Ser37/Thr41 and its subsequent degradation in human retinal pigment epithelial (RPE) cells. Wnt5a decreased the levels of vascular endothelial growth factor (VEGF), tumor necrosis factor-α(TNF-α), and nuclear factor-κB (NF-κB), which was up-regulated by Wnt3a. Furthermore, Wnt5a increased E-cadherin expression and decreased cell migration by down-regulating Snail expression, thereby abrogating the Wnt3a-induced epithelial-mesenchymal transition (EMT) in human RPE cells. Our findings suggest that Wnt5a suppresses the pathogenic effects of canonical Wnt signaling in human RPE cells by promoting β-catenin phosphorylation and degradation. Therefore, Wnt5a has significant therapeutic potential for the treatment of AMD. [BMB Reports 2015; 48(9): 525-530]
Collapse
Affiliation(s)
- Joo-Hyun Kim
- Department of Bio and Fermentation Convergence Technology, Kookmin University, Seoul 02707, Korea
| | - Seoyoung Park
- Department of Bio and Fermentation Convergence Technology, Kookmin University, Seoul 02707, Korea
| | - Hyewon Chung
- Department of Opthalmology, Konkuk University School of Medicine, Seoul 05066, Korea
| | - Sangtaek Oh
- Department of Bio and Fermentation Convergence Technology, Kookmin University, Seoul 02707, Korea
| |
Collapse
|
28
|
Ishikawa K, Sreekumar PG, Spee C, Nazari H, Zhu D, Kannan R, Hinton DR. αB-Crystallin Regulates Subretinal Fibrosis by Modulation of Epithelial-Mesenchymal Transition. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:859-73. [PMID: 26878210 PMCID: PMC4822331 DOI: 10.1016/j.ajpath.2015.11.014] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Revised: 10/20/2015] [Accepted: 11/19/2015] [Indexed: 01/18/2023]
Abstract
Subretinal fibrosis is an end stage of neovascular age-related macular degeneration, characterized by fibrous membrane formation after choroidal neovascularization. An initial step of the pathogenesis is an epithelial-mesenchymal transition (EMT) of retinal pigment epithelium cells. αB-crystallin plays multiple roles in age-related macular degeneration, including cytoprotection and angiogenesis. However, the role of αB-crystallin in subretinal EMT and fibrosis is unknown. Herein, we showed attenuation of subretinal fibrosis after regression of laser-induced choroidal neovascularization and a decrease in mesenchymal retinal pigment epithelium cells in αB-crystallin knockout mice compared with wild-type mice. αB-crystallin was prominently expressed in subretinal fibrotic lesions in mice. In vitro, overexpression of αB-crystallin induced EMT, whereas suppression of αB-crystallin induced a mesenchymal-epithelial transition. Transforming growth factor-β2-induced EMT was further enhanced by overexpression of αB-crystallin but was inhibited by suppression of αB-crystallin. Silencing of αB-crystallin inhibited multiple fibrotic processes, including cell proliferation, migration, and fibronectin production. Bone morphogenetic protein 4 up-regulated αB-crystallin, and its EMT induction was inhibited by knockdown of αB-crystallin. Furthermore, inhibition of αB-crystallin enhanced monotetraubiquitination of SMAD4, which can impair its nuclear localization. Overexpression of αB-crystallin enhanced nuclear translocation and accumulation of SMAD4 and SMAD5. Thus, αB-crystallin is an important regulator of EMT, acting as a molecular chaperone for SMAD4 and as its potential therapeutic target for preventing subretinal fibrosis development in neovascular age-related macular degeneration.
Collapse
Affiliation(s)
- Keijiro Ishikawa
- Arnold and Mabel Beckman Macular Research Center, Doheny Eye Institute, Los Angeles, California; Department of Ophthalmology, Keck School of Medicine of the University of Southern California, Los Angeles, California
| | | | - Christine Spee
- Department of Ophthalmology, Keck School of Medicine of the University of Southern California, Los Angeles, California
| | - Hossein Nazari
- Department of Ophthalmology, Keck School of Medicine of the University of Southern California, Los Angeles, California
| | - Danhong Zhu
- Department of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, California
| | - Ram Kannan
- Arnold and Mabel Beckman Macular Research Center, Doheny Eye Institute, Los Angeles, California
| | - David R Hinton
- Department of Ophthalmology, Keck School of Medicine of the University of Southern California, Los Angeles, California; Department of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, California.
| |
Collapse
|
29
|
Yang S, Yao H, Li M, Li H, Wang F. Long Non-Coding RNA MALAT1 Mediates Transforming Growth Factor Beta1-Induced Epithelial-Mesenchymal Transition of Retinal Pigment Epithelial Cells. PLoS One 2016; 11:e0152687. [PMID: 27019196 PMCID: PMC4809592 DOI: 10.1371/journal.pone.0152687] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 03/17/2016] [Indexed: 11/19/2022] Open
Abstract
PURPOSE To study the role of long non-coding RNA (lncRNA) MALAT1 in transforming growth factor beta 1 (TGF-β1)-induced epithelial-mesenchymal transition (EMT) of retinal pigment epithelial (RPE) cells. METHODS ARPE-19 cells were cultured and exposed to TGF-β1. The EMT of APRE-19 cells is confirmed by morphological change, as well as the increased expression of alpha-smooth muscle actin (αSMA) and fibronectin, and the down-regulation of E-cadherin and Zona occludin-1(ZO-1) at both mRNA and protein levels. The expression of lncRNA MALAT1 in RPE cells were detected by quantitative real-time PCR. Knockdown of MALAT1 was achieved by transfecting a small interfering RNA (SiRNA). The effect of inhibition of MALAT1 on EMT, migration, proliferation, and TGFβ signalings were observed. MALAT1 expression was also detected in primary RPE cells incubated with proliferative vitreoretinopathy (PVR) vitreous samples. RESULTS The expression of MALAT1 is significantly increased in RPE cells incubated with TGFβ1. MALAT1 silencing attenuates TGFβ1-induced EMT, migration, and proliferation of RPE cells, at least partially through activating Smad2/3 signaling. MALAT1 is also significantly increased in primary RPE cells incubated with PVR vitreous samples. CONCLUSION LncRNA MALAT1 is involved in TGFβ1-induced EMT of human RPE cells and provides new understandings for the pathogenesis of PVR.
Collapse
Affiliation(s)
- Shuai Yang
- Department of Ophthalmology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Haipei Yao
- Department of Ophthalmology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Min Li
- Department of Ophthalmology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Hui Li
- Department of Ophthalmology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Fang Wang
- Department of Ophthalmology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, 200072, China
- * E-mail:
| |
Collapse
|
30
|
Osorio LA, Farfán NM, Castellón EA, Contreras HR. SNAIL transcription factor increases the motility and invasive capacity of prostate cancer cells. Mol Med Rep 2015; 13:778-86. [PMID: 26648419 PMCID: PMC4686115 DOI: 10.3892/mmr.2015.4585] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 10/28/2015] [Indexed: 11/18/2022] Open
Abstract
The incidence and mortality rates of prostate cancer (PCa) are increasing, and PCa is almost the second-leading cause of cancer-associated mortality in men. During tumor progression, epithelial cells decrease the number of adhesion molecules, change their polarity and position, rearrange their cytoskeleton and increase their migratory and invasive capacities. These changes are known under the concept of epithelial-mesenchymal transition (EMT). EMT is characterized by an upregulation of certain transcription factors, including SNAIL1, which represses genes that are characteristic of an epithelial phenotype, including E-cadherin, and indirectly increase the expression levels of genes, which are associated with the mesenchymal phenotype. It has been suggested that the transcription factor, SNAIL1, decreases the proliferation and increases the migratory and invasive capacities of PCa cell lines. The present study was performed using LNCaP and PC3 cell lines, in which the expression levels of SNAIL1 were increased or silenced through the use of lentiviral vectors. The expression levels of EMT markers were quantified using reverse transcription-quantitative polymerase chain reaction and western blot analysis. In addition, cell survival was analyzed using an MTS assay; cell proliferation was examined using an antibody targeting Ki-67; migration on plates with 8 µm pores to allow the passage of cells; and invasiveness was analyzed using a membrane chamber covered in dried basement membrane matrix solution. The levels of apoptosis were determined using a Caspase 3/7 assay containing a substrate modified by caspases 3 and 7. The results demonstrated that the overexpression and silencing of SNAIL1 decreased cell proliferation and survival. However, the overexpression of SNAIL1 decreased apoptosis, compared with cells with the SNAIL1-silenced cells, in which cell apoptosis increased. The migration and invasive capacities increased in the cells overexpressing SNAIL1, and decreased when SNAIL1 was silenced. In conclusion, PCa cells overexpressing SNAIL1 exhibited characteristics of an EMT phenotype, whereas the silencing of the SNAIL1 transcriptional repressor promoted an epithelial-like phenotype, with decreased migration and invasion, characteristic of mesenchymal cells.
Collapse
Affiliation(s)
- Luis A Osorio
- Physiology and Biophysics Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago 8389100, Chile
| | - Nancy M Farfán
- Physiology and Biophysics Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago 8389100, Chile
| | - Enrique A Castellón
- Physiology and Biophysics Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago 8389100, Chile
| | - Héctor R Contreras
- Physiology and Biophysics Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago 8389100, Chile
| |
Collapse
|
31
|
Abstract
Results of the present study support ocular epithelia-specific LOXL1 functions in exfoliation glaucoma that may include both dysregulated extracellular matrix cross-linking activity and cellular mechanisms involving a role for LOXL1, in direct interaction with Snail1, in promoting epithelial to mesenchymal transition and a potential shift towards fibrogenic epithelial cell phenotypes.
Collapse
|
32
|
Combined silencing of TGF-β2 and Snail genes inhibit epithelial-mesenchymal transition of retinal pigment epithelial cells under hypoxia. Graefes Arch Clin Exp Ophthalmol 2015; 253:875-84. [PMID: 25875044 DOI: 10.1007/s00417-014-2922-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 12/14/2014] [Accepted: 12/27/2014] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The formation of scar-like fibrous tissue in age-related macular degeneration (AMD) is associated with hypoxia. Under hypoxia, retinal pigment epithelial (RPE) cells can secret more transforming growth factor-β2 (TGF-β2), which is determined to induce epithelial-mesenchymal transition (EMT) at certain concentrations. Whether hypoxia can induce EMT by stimulating RPE cell line secrets TGF-β2 or not remains unknown. To gain a better understanding of the signaling mechanisms of fibrosis in AMD under hypoxic conditions, we investigated EMT in retinal pigment epithelial (RPE) cells and the effect of TGF-β2 and Snail in this process. METHODS Human RPE cell line (ARPE-19) was incubated with 5 % O2 for different periods of time. The expression of N-cadherin, α-smooth muscle actin (α-SMA), TGF-β2 , and Snail were determined by Western blot and real-time PCR. Cell proliferation was assessed by CCK8 kit. RNA interference was used for multi-gene silencing of TGF-β2 and Snail genes. RESULTS N-cadherin was decreased and mesenchymal cell marker α-SMA was increased after the ARPE-19 cell line was incubated with 5 % O2. Meanwhile, the proliferation capability of the cell line was increased. TGF-β2 and Snail expression were increased in a time-dependent manner under hypoxia. After multi-silencing TGF-β2 and Snail genes, N-cadherin was increased and α-SMA was reduced. Meanwhile, the proliferation of the cell line was suppressed. CONCLUSIONS Under hypoxic conditions, RPE cells undergo EMT. Endogenic TGF-β2 and Snail are involved in this process. Furthermore, knockdown of both TGF-β2 and Snail inhibited EMT to a greater extent than knockdown of either gene individually.
Collapse
|