1
|
Galindo-Cabello N, Sobas-Abad EM, Lapresa R, Agulla J, Almeida Á, López A, Pastor JC, Pastor-Idoate S, Usategui-Martín R. The TP53 Arg72Pro polymorphism predicts visual and neurodegenerative outcomes in retinal detachment. Cell Death Dis 2025; 16:415. [PMID: 40419469 DOI: 10.1038/s41419-025-07739-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 05/03/2025] [Accepted: 05/15/2025] [Indexed: 05/28/2025]
Abstract
Retinal detachment (RD) separates the retina from the retinal epithelium, causing photoreceptor apoptosis and irreversible vision loss. Even with successful surgical reattachment, complete visual recovery is not guaranteed. The TP53 Arg72Pro polymorphism, implicated in apoptosis, has emerged as a potential predictor of RD outcomes. We investigated the impact of the Arg72Pro polymorphism on retinal neurodegeneration and functional recovery in patients. The underlying mechanisms were analyzed in a humanized TP53 Arg72Pro RD mouse model. In a cohort of 180 patients, carriers of the Pro allele exhibited decreased apoptotic gene expression and improved visual recovery. Complementary findings in mice revealed that the Pro variant preserved photoreceptor integrity and reduced apoptosis rates following RD. Our findings highlight the potential of this TP53 polymorphism as a biomarker for RD outcomes and a tool for tailoring therapies. This study underscores the importance of integrating genetic profiling into personalized medicine approaches to improve recovery of RD patients' visual outcomes.
Collapse
Affiliation(s)
- Nadia Galindo-Cabello
- Unit of Excellence Institute of Applied Ophthalmobiology (IOBA), University of Valladolid, Valladolid, Spain
- Department of Cell Biology, Genetics, Histology and Pharmacology, Faculty of Medicine, University of Valladolid, Valladolid, Spain
- Network of Inflammatory Diseases- Networks of Cooperative Research Oriented to Health Results (RICORS), Carlos III National Institute of Health, Madrid, Spain
| | - Eva M Sobas-Abad
- Unit of Excellence Institute of Applied Ophthalmobiology (IOBA), University of Valladolid, Valladolid, Spain
- Network of Inflammatory Diseases- Networks of Cooperative Research Oriented to Health Results (RICORS), Carlos III National Institute of Health, Madrid, Spain
- School of Nursing, University of Valladolid, Valladolid, Spain
| | - Rebeca Lapresa
- Institute of Biomedical Research of Salamanca (IBSAL), University Hospital of Salamanca, University of Salamanca, CSIC, Salamanca, Spain
- Institute of Functional Biology and Genomics, CSIC, University of Salamanca, Salamanca, Spain
| | - Jesús Agulla
- Institute of Biomedical Research of Salamanca (IBSAL), University Hospital of Salamanca, University of Salamanca, CSIC, Salamanca, Spain
- Institute of Functional Biology and Genomics, CSIC, University of Salamanca, Salamanca, Spain
| | - Ángeles Almeida
- Institute of Biomedical Research of Salamanca (IBSAL), University Hospital of Salamanca, University of Salamanca, CSIC, Salamanca, Spain
- Institute of Functional Biology and Genomics, CSIC, University of Salamanca, Salamanca, Spain
| | - Antonio López
- Unit of Excellence Institute of Applied Ophthalmobiology (IOBA), University of Valladolid, Valladolid, Spain
| | - José Carlos Pastor
- Unit of Excellence Institute of Applied Ophthalmobiology (IOBA), University of Valladolid, Valladolid, Spain
| | - Salvador Pastor-Idoate
- Unit of Excellence Institute of Applied Ophthalmobiology (IOBA), University of Valladolid, Valladolid, Spain.
- Network of Inflammatory Diseases- Networks of Cooperative Research Oriented to Health Results (RICORS), Carlos III National Institute of Health, Madrid, Spain.
- Department of Ophthalmology, University Clinical Hospital of Valladolid, Valladolid, Spain.
| | - Ricardo Usategui-Martín
- Unit of Excellence Institute of Applied Ophthalmobiology (IOBA), University of Valladolid, Valladolid, Spain.
- Department of Cell Biology, Genetics, Histology and Pharmacology, Faculty of Medicine, University of Valladolid, Valladolid, Spain.
- Network of Inflammatory Diseases- Networks of Cooperative Research Oriented to Health Results (RICORS), Carlos III National Institute of Health, Madrid, Spain.
| |
Collapse
|
2
|
Han H, Yang Y, Han Z, Wang L, Dong L, Qi H, Liu B, Tian J, Vanhaesebroeck B, Kazlauskas A, Zhang G, Zhang S, Lei H. NFκB-Mediated Expression of Phosphoinositide 3-Kinase δ Is Critical for Mesenchymal Transition in Retinal Pigment Epithelial Cells. Cells 2023; 12:207. [PMID: 36672142 PMCID: PMC9857235 DOI: 10.3390/cells12020207] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/11/2022] [Accepted: 12/12/2022] [Indexed: 01/05/2023] Open
Abstract
Epithelial mesenchymal transition (EMT) plays a vital role in a variety of human diseases including proliferative vitreoretinopathy (PVR), in which retinal pigment epithelial (RPE) cells play a key part. Transcriptomic analysis showed that the phosphoinositide 3-kinase (PI3K)/Akt signaling pathway was up-regulated in human RPE cells upon treatment with transforming growth factor (TGF)-β2, a multifunctional cytokine associated with clinical PVR. Stimulation of human RPE cells with TGF-β2 induced expression of p110δ (the catalytic subunit of PI3Kδ) and activation of NFκB/p65. CRISPR-Cas9-mediated depletion of p110δ or NFκB/p65 suppressed TGF-β2-induced fibronectin expression and activation of Akt as well as migration of these cells. Intriguingly, abrogating expression of NFκB/p65 also blocked TGF-β2-induced expression of p110δ, and luciferase reporter assay indicated that TGF-β2 induced NFκB/p65 binding to the promoter of the PIK3CD that encodes p110δ. These data reveal that NFκB/p65-mediated expression of PI3Kδ is essential in human RPE cells for TGF-β2-induced EMT, uncovering hindrance of TGF-β2-induced expression of p110δ as a novel approach to inhibit PVR.
Collapse
Affiliation(s)
- Haote Han
- Institute of Basic Medicine and Cancer, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou 100864, China
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA 02114, USA
- Department of Ophthalmology, Harvard Medical School, Boston, MA 02115, USA
| | - Yanhui Yang
- Ningxia Key Laboratory of Prevention and Control of Common Infectious Diseases, The School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750101, China
| | - Zhuo Han
- Institute of Basic Medicine and Cancer, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou 100864, China
| | - Luping Wang
- Institute of Basic Medicine and Cancer, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou 100864, China
| | - Lijun Dong
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, Shenzhen 518000, China
| | - Hui Qi
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, Shenzhen 518000, China
| | - Bing Liu
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA 02114, USA
- Department of Ophthalmology, Harvard Medical School, Boston, MA 02115, USA
- Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou 510180, China
| | - Jingkui Tian
- Institute of Basic Medicine and Cancer, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou 100864, China
| | | | - Andrius Kazlauskas
- Department of Ophthalmology and Visual Sciences, Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Guoming Zhang
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, Shenzhen 518000, China
| | - Shaochong Zhang
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, Shenzhen 518000, China
| | - Hetian Lei
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, Shenzhen 518000, China
| |
Collapse
|
3
|
Wu W, Ma G, Qi H, Dong L, Chen F, Wang Y, Mao X, Guo X, Cui J, Matsubara JA, Vanhaesebroeck B, Yan X, Zhao G, Zhang S, Lei H. Genome Editing of Pik3cd Impedes Abnormal Retinal Angiogenesis. Hum Gene Ther 2023; 34:30-41. [PMID: 36515172 DOI: 10.1089/hum.2022.079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Abnormal angiogenesis is associated with myriad human diseases, including proliferative diabetic retinopathy (PDR). Signaling transduction through phosphoinositide 3-kinases (PI3Ks) plays a critical role in angiogenesis. Herein, we showed that p110δ, the catalytic subunit of PI3Kδ, was highly expressed in pathological retinal vascular endothelial cells (ECs) in a mouse model of oxygen-induced retinopathy (OIR) and in fibrovascular membranes from patients with PDR. To explore novel intervention with PI3Kδ expression, we developed a recombinant dual adeno-associated viral (rAAV) system for delivering CRISPR/Cas9 in which Streptococcus pyogenes (Sp) Cas9 expression was driven by an endothelial specific promoter of the intercellular adhesion molecule 2 (pICAM2) to edit genomic Pik3cd, the gene encoding p110δ. We then demonstrated that infection of cultured mouse vascular ECs with the dual rAAV1s of rAAV1-pICAM2-SpCas9 and rAAV1-SpGuide targeting genomic Pik3cd resulted in 80% DNA insertion/deletion in the locus of genomic Pik3cd and 70% depletion of p110δ expression. Furthermore, we showed that in the mouse model of OIR editing retinal Pik3cd with the dual rAAV1s resulted in not only a significant decrease in p110δ expression, and Akt activation, but also a dramatic reduction in pathological retinal angiogenesis. These findings reveal that Pik3cd editing is a novel approach to treating abnormal retinal angiogenesis.
Collapse
Affiliation(s)
- Wenyi Wu
- Department of Ophthalmology, Hunan Key Laboratory of Ophthalmology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.,Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, USA.,Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, USA
| | - Gaoen Ma
- Department of Ophthalmology, The Third Affiliated Hospital of Xinxiang Medical University, Eye Hospital of Xinxiang Medical University, Xinxiang, China
| | - Hui Qi
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, Shenzhen, China
| | - Lijun Dong
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, Shenzhen, China
| | - Fang Chen
- Huan Key Laboratory of Molecular Precision Medicine, Xiangya Hospital and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Yun Wang
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, Shenzhen, China
| | - Xingxing Mao
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, Shenzhen, China
| | - Xiaoqing Guo
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, USA.,Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, USA
| | - Jing Cui
- The University of British Columbia, Vancouver, Canada
| | | | | | - Xiaohe Yan
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, Shenzhen, China
| | - Guoming Zhao
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, Shenzhen, China
| | - Shaochong Zhang
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, Shenzhen, China
| | - Hetian Lei
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, Shenzhen, China
| |
Collapse
|
4
|
Dong L, Han H, Huang X, Ma G, Fang D, Qi H, Han Z, Wang L, Tian J, Vanhaesebroeck B, Zhang G, Zhang S, Lei H. Idelalisib inhibits experimental proliferative vitroretinopathy. J Transl Med 2022; 102:1296-1303. [PMID: 35854067 DOI: 10.1038/s41374-022-00822-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 06/16/2022] [Accepted: 06/21/2022] [Indexed: 11/09/2022] Open
Abstract
Proliferative vitreoretinopathy (PVR) is a fibrotic eye disease that develops after rhegmatogenous retinal detachment surgery and open-globe traumatic injury. Idelalisib is a specific inhibitor of phosphoinositide 3-kinase (PI3K) δ. While PI3Kδ is primarily expressed in leukocytes, its expression is also considerably high in retinal pigment epithelial (RPE) cells, which play a crucial part in the PVR pathogenesis. Herein we show that GeoMx Digital Spatial Profiling uncovered strong expression of fibronectin in RPE cells within epiretinal membranes from patients with PVR, and that idelalisib (10 μM) inhibited Akt activation, fibronectin expression and collagen gel contraction induced by transforming growth factor (TGF)-β2 in human RPE cells. Furthermore, we discovered that idelalisib at a vitreal concentration of 10 μM, a non-toxic dose to the retina, prevented experimental PVR induced by intravitreally injected RPE cells in rabbits assessed by experienced ophthalmologists using an indirect ophthalmoscope plus a + 30 D fundus lens, electroretinography, optical coherence tomography and histological analysis. These data suggested idelalisib could be harnessed for preventing patients from PVR.
Collapse
Affiliation(s)
- Lijun Dong
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, Shenzhen, China
| | - Haote Han
- Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China.,Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA, USA.,Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Xionggao Huang
- Department of Ophthalmology, the First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Gaoen Ma
- Department of Ophthalmology, the third Hospital of Xinxiang Medical University, Xinxiang, China
| | - Dong Fang
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, Shenzhen, China
| | - Hui Qi
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, Shenzhen, China
| | - Zhuo Han
- Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
| | - Luping Wang
- Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
| | - Jingkui Tian
- Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
| | | | - Guoming Zhang
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, Shenzhen, China
| | - Shaochong Zhang
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, Shenzhen, China.
| | - Hetian Lei
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, Shenzhen, China.
| |
Collapse
|
5
|
Moledina M, Charteris DG, Chandra A. The Genetic Architecture of Non-Syndromic Rhegmatogenous Retinal Detachment. Genes (Basel) 2022; 13:genes13091675. [PMID: 36140841 PMCID: PMC9498391 DOI: 10.3390/genes13091675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 09/14/2022] [Accepted: 09/16/2022] [Indexed: 11/16/2022] Open
Abstract
Rhegmatogenous retinal detachment (RRD) is the most common form of retinal detachment (RD), affecting 1 in 10,000 patients per year. The condition has significant ocular morbidity, with a sizeable proportion of patients obtaining poor visual outcomes. Despite this, the genetics underpinning Idiopathic Retinal Detachment (IRD) remain poorly understood; this is likely due to small sample sizes in relevant studies. The majority of research pertains to the well-characterised Mende lian syndromes, such as Sticklers and Wagners, associated with RRD. Nevertheless, in recent years, there has been an increasing body of literature identifying the common genetic mutations and mechanisms associated with IRD. Several recent Genomic Wide Association Studies (GWAS) studies have identified a number of genetic loci related to the development of IRD. Our review aims to provide an up-to-date summary of the significant genetic mechanisms and associations of Idiopathic RRD.
Collapse
Affiliation(s)
- Malik Moledina
- Department of Ophthalmology, Southend University Hospital, Mid & South Essex NHS Foundation Trust, Southend-on-Sea SS0 0RY, UK
| | - David G. Charteris
- Institute of Ophthalmology, University College, London EC1V 9EL, UK
- Vitreoretinal Unit, Moorfields Eye Hospital NHS Foundation Trust, London EC1V 2PD, UK
| | - Aman Chandra
- Department of Ophthalmology, Southend University Hospital, Mid & South Essex NHS Foundation Trust, Southend-on-Sea SS0 0RY, UK
- School of Medicine, Anglia Ruskin University, Chelmsford CM1 1SQ, UK
- Correspondence: ; Tel.: +44-7914-817445
| |
Collapse
|
6
|
Zauli G, AlHilali S, Al-Swailem S, Secchiero P, Voltan R. Therapeutic potential of the MDM2 inhibitor Nutlin-3 in counteracting SARS-CoV-2 infection of the eye through p53 activation. Front Med (Lausanne) 2022; 9:902713. [PMID: 35911386 PMCID: PMC9329687 DOI: 10.3389/fmed.2022.902713] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 06/27/2022] [Indexed: 01/08/2023] Open
Abstract
Starting from the beginning of the severe acute respiratory syndrome Coronavirus-2 (SARS-CoV-2) global pandemic, most of the published data has concentrated on the respiratory signs and symptoms of Covid-19 infection, underestimating the presence and importance of ocular manifestations, such as conjunctivitis, usually reported in SARS-CoV-2 infected patients. With the present review we intend to resume the ocular involvement in SARS-CoV-2 infection and the recent discoveries about the different cell types and tissues of the eye that can be directly infected by SARS-CoV-2 and propagate the infection. Moreover, reviewing literature data about p53 expression in normal and diseased eye tissues, we hypothesize that the pleiotropic protein p53 present at high levels in cornea, conjunctiva and tear film might play a protective role against SARS-CoV-2 infection. Since p53 can be easily up-regulated by using small molecule non-genotoxic inhibitors of MDM2, we propose that topical use of Nutlin-3, the prototype member of MDM2 inhibitors, might protect the anterior surface of the eye from SARS-CoV-2 infection, reducing the spreading of the virus.
Collapse
Affiliation(s)
- Giorgio Zauli
- Research Department, King Khaled Eye Specialistic Hospital, Riyadh, Saudi Arabia
- *Correspondence: Giorgio Zauli
| | - Sara AlHilali
- Research Department, King Khaled Eye Specialistic Hospital, Riyadh, Saudi Arabia
| | - Samar Al-Swailem
- Research Department, King Khaled Eye Specialistic Hospital, Riyadh, Saudi Arabia
| | - Paola Secchiero
- Department of Translational Medicine and LTTA Centre, University of Ferrara, Ferrara, Italy
| | - Rebecca Voltan
- Department of Environmental and Prevention Sciences and LTTA Centre, University of Ferrara, Ferrara, Italy
| |
Collapse
|
7
|
Jiang H, Yan B, Meng Z, Zhang L, Lei H, Luo J. The MDM2 Single-Nucleotide Polymorphism T309G Is Associated With the Development of Epimacular Membranes. Front Cell Dev Biol 2022; 10:841660. [PMID: 35359434 PMCID: PMC8963840 DOI: 10.3389/fcell.2022.841660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/15/2022] [Indexed: 11/13/2022] Open
Abstract
Purpose: To investigate the role of the mouse double minute 2 (MDM2) gene single-nucleotide polymorphism (SNP) T309G in the development of epimacular membranes (EMMs) by analyzing the genotype distribution and consistency of the polymorphism in paired membrane-blood samples. Methods: This was a cross–sectional genetic association study of patients with proliferative vitreoretinopathy (PVR) or EMMs. PVR membranes (PVRMs), internal limiting membranes (ILMs) (PVR-ILMs) and blood samples (PVR-blood) from patients with PVR, and EMMs, EMM-ILMs and EMM-blood from patients with EMMs were collected. The genotype of all samples was determined by Sanger sequencing. Sex composition, mean age, the genotype distribution of MDM2 T309G, the allelic frequency of the MDM2 SNP309 G allele (% G) and the somatic mutation rate at the MDM2 T309G locus (% M) were analyzed and compared. The PVR and healthy Chinese donor groups were used as controls for different comparisons. Results: The EMM group of 62 patients was older than the PVR group of 61 patients by an average of 8.87 years (p < 0.0001), but the two groups were statistically similar in the sex composition (p = 0.1754). Importantly, G allele carriers were at a higher risk of developing EMMs than non-G allele carriers (p = 0.0479; OR = 2.047). Moreover, EMM-blood exhibited a significantly higher % G than blood samples from healthy Chinese donors (EMM-blood: 56.78%, donors: 45.61%; p = 0.0256; OR = 1.567). Regarding membrane-blood consistency, % M was significantly different between PVRMs and EMMs (PVRMs: 2.63%, EMMs: 21.57%; p = 0.0097; OR = 10.18) but not between different types of ILMs (PVR-ILMs: 18.18%, EMM-ILMs: 29.17%; p = 0.6855). Furthermore, EMMs (p = 0.0053; OR = 8.250) and EMM-ILMs (p = 0.0233; OR = 14.40) from patients with preoperative macular holes were more predisposed toward somatic mutations at the MDM2 T309G locus than those from patients without preoperative macular holes. Conclusions:MDM2 T309G is associated with the development of EMMs. Herein, the MDM2 SNP309 G allele is first reported as an associated factor of EMMs in a Chinese population. In addition, EMMs and ILMs are genetically unstable at the MDM2 T309G locus, especially when complicated with preoperative macular holes.
Collapse
Affiliation(s)
- Heng Jiang
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Bin Yan
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Zhishang Meng
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Lusi Zhang
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Hetian Lei
- Shenzhen Eye Institute, Shenzhen Eye Hospital, Jinan University, Shenzhen, China
- *Correspondence: Hetian Lei, , Jing Luo,
| | - Jing Luo
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Hetian Lei, , Jing Luo,
| |
Collapse
|
8
|
The roles of mouse double minute 2 (MDM2) oncoprotein in ocular diseases: A review. Exp Eye Res 2022; 217:108910. [PMID: 34998788 DOI: 10.1016/j.exer.2021.108910] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 12/03/2021] [Accepted: 12/21/2021] [Indexed: 12/19/2022]
Abstract
Mouse double minute 2 (MDM2), an E3 ubiquitin ligase and the primary negative regulator of the tumor suppressor p53, cooperates with its structural homolog MDM4/MDMX to control intracellular p53 level. In turn, overexpression of p53 upregulates and forms an autoregulatory feedback loop with MDM2. The MDM2-p53 axis plays a pivotal role in modulating cell cycle control and apoptosis. MDM2 itself is regulated by the PI3K-AKT and RB-E2F-ARF pathways. While amplification of the MDM2 gene or overexpression of MDM2 (due to MDM2 SNP T309G, for instance) is associated with various malignancies, numerous studies have shown that MDM2/p53 alterations may also play a part in the pathogenetic process of certain ocular disorders (Fig. 1). These include cancers (retinoblastoma, uveal melanoma), fibrocellular proliferative diseases (proliferative vitreoretinopathy, pterygium), neovascular diseases, degenerative diseases (cataract, primary open-angle glaucoma, age-related macular degeneration) and infectious/inflammatory diseases (trachoma, uveitis). In addition, MDM2 is implicated in retinogenesis and regeneration after optic nerve injury. Anti-MDM2 therapy has shown potential as a novel approach to treating these diseases. Despite major safety concerns, there are high expectations for the clinical value of reformative MDM2 inhibitors. This review summarizes important findings about the role of MDM2 in ocular pathologies and provides an overview of recent advances in treating these diseases with anti-MDM2 therapies.
Collapse
|
9
|
Han H, Yang Y, Liu B, Tian J, Dong L, Qi H, Zhu W, Wang J, Lei H. Chalcomoracin prevents vitreous-induced activation of AKT and migration of retinal pigment epithelial cells. J Cell Mol Med 2021; 25:9102-9111. [PMID: 34432370 PMCID: PMC8500972 DOI: 10.1111/jcmm.16590] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 02/19/2021] [Accepted: 04/15/2021] [Indexed: 01/17/2023] Open
Abstract
Retinal pigment epithelial (RPE) cells are the major cell type in the epi- or sub-retinal membranes in the pathogenesis of proliferative vitreoretinopathy (PVR), which is a blinding fibrotic eye disease and still short of effective medicine. The purpose of this study is to demonstrate whether Chalocomoracin (CMR), a novel purified compound from fungus-infected mulberry leaves, is able to inhibit vitreous-induced signalling events and cellular responses intrinsic to PVR. Our studies have revealed that the CMR IC50 for ARPE-19 cells is 35.5 μmol/L at 72 hours, and that 5 μmol/L CMR inhibits vitreous-induced Akt activation and p53 suppression; in addition, we have discovered that this chemical effectively blocks vitreous-stimulated proliferation, migration and contraction of ARPE-19 cells, suggesting that CMR is a promising PVR prophylactic.
Collapse
Affiliation(s)
- Haote Han
- Institute of Cancer and Basic MedicineChinese Academy of SciencesZhejiang Cancer HospitalCancer Hospital of the University of Chinese Academy of SciencesHangzhouChina
- College of Biomedical Engineering & Instrument ScienceZhejiang UniversityHangzhouChina
- Schepens Eye Research Institute of Massachusetts Eye and EarBostonMAUSA
- Department of OphthalmologyHarvard Medical SchoolBostonMAUSA
| | - Yanhui Yang
- School of Basic Medical SciencesNingxia Medical UniversityYinchuanChina
| | - Bing Liu
- Guangzhou Women and Children's Medical CenterGuangzhou Medical UniversityGuangzhouChina
| | - Jingkui Tian
- Institute of Cancer and Basic MedicineChinese Academy of SciencesZhejiang Cancer HospitalCancer Hospital of the University of Chinese Academy of SciencesHangzhouChina
- College of Biomedical Engineering & Instrument ScienceZhejiang UniversityHangzhouChina
| | - Lijun Dong
- Shenzhen Eye HospitalShenzhen Eye InstituteJinan UniversityShenzhenChina
| | - Hui Qi
- Shenzhen Eye HospitalShenzhen Eye InstituteJinan UniversityShenzhenChina
| | - Wei Zhu
- Institute of Cancer and Basic MedicineChinese Academy of SciencesZhejiang Cancer HospitalCancer Hospital of the University of Chinese Academy of SciencesHangzhouChina
- College of Biomedical Engineering & Instrument ScienceZhejiang UniversityHangzhouChina
| | - Jiantao Wang
- Shenzhen Eye HospitalShenzhen Eye InstituteJinan UniversityShenzhenChina
| | - Hetian Lei
- Shenzhen Eye HospitalShenzhen Eye InstituteJinan UniversityShenzhenChina
| |
Collapse
|
10
|
Yang Y, Huang X, Ma G, Cui J, Matsubara JA, Kazlauskas A, Zhao J, Wang J, Lei H. PDGFRβ plays an essential role in patient vitreous-stimulated contraction of retinal pigment epithelial cells from epiretinal membranes. Exp Eye Res 2020; 197:108116. [PMID: 32561481 DOI: 10.1016/j.exer.2020.108116] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 05/28/2020] [Accepted: 06/08/2020] [Indexed: 12/28/2022]
Abstract
Platelet-derived growth factor (PDGF) is associated with clinical proliferative vitreoretinopathy (PVR), which is characterized by formation of sub- or epi-retinal membranes that consist of cells including retinal pigment epithelial (RPE) cells and extracellular matrix. RPE cells play an important role in PVR pathogenesis. Previous findings indicated that PDGF receptor (PDGFR)α was essential in experimental PVR induced by fibroblasts. In RPE cells derived from epiretinal membranes from patients with PVR (RPEMs), Akt was activated by PDGF-B but not PDGF-A, which suggested that PDGFRβ was the predominant PDGFR isoform expressed in RPEMs. Indeed, CRISPR/Cas9-mediated depletion of PDGFRβ in RPEMs attenuated patient vitreous-induced Akt activation and cellular responses intrinsic to PVR including cell proliferation, migration, and contraction. We conclude that PDGFRβ appears to be the PVR relevant PDGFR isoform in RPEMs.
Collapse
Affiliation(s)
- Yanhui Yang
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia, PR China; Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Xionggao Huang
- Department of Ophthalmology, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan Province, PR China
| | - Gaoen Ma
- Department of Ophthalmology, The Third Hospital of Xinxiang Medical University, Xinxiang, Henan Province, PR China
| | - Jing Cui
- The University of British Columbia, Canada
| | | | - Andrius Kazlauskas
- Department of Ophthalmology, University of Illinois at Chicago, Chicago, USA
| | - Jun Zhao
- Shenzhen Eye Hospital, Shenzhen Eye Institute, Jinan University, Shenzhen, Guangdong Province, PR China
| | - Jiantao Wang
- Shenzhen Eye Hospital, Shenzhen Eye Institute, Jinan University, Shenzhen, Guangdong Province, PR China.
| | - Hetian Lei
- Shenzhen Eye Hospital, Shenzhen Eye Institute, Jinan University, Shenzhen, Guangdong Province, PR China.
| |
Collapse
|
11
|
Wu W, Zhou G, Han H, Huang X, Jiang H, Mukai S, Kazlauskas A, Cui J, Matsubara JA, Vanhaesebroeck B, Xia X, Wang J, Lei H. PI3Kδ as a Novel Therapeutic Target in Pathological Angiogenesis. Diabetes 2020; 69:736-748. [PMID: 31915155 PMCID: PMC7085248 DOI: 10.2337/db19-0713] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Accepted: 12/25/2019] [Indexed: 12/12/2022]
Abstract
Diabetic retinopathy is the most common microvascular complication of diabetes, and in the advanced diabetic retinopathy appear vitreal fibrovascular membranes that consist of a variety of cells, including vascular endothelial cells (ECs). New therapeutic approaches for this diabetic complication are urgently needed. Here, we report that in cultured human retinal microvascular ECs, high glucose induced expression of p110δ, which was also expressed in ECs of fibrovascular membranes from patients with diabetes. This catalytic subunit of a receptor-regulated PI3K isoform δ is known to be highly enriched in leukocytes. Using genetic and pharmacological approaches, we show that p110δ activity in cultured ECs controls Akt activation, cell proliferation, migration, and tube formation induced by vascular endothelial growth factor, basic fibroblast growth factor, and epidermal growth factor. Using a mouse model of oxygen-induced retinopathy, p110δ inactivation was found to attenuate pathological retinal angiogenesis. p110δ inhibitors have been approved for use in human B-cell malignancies. Our data suggest that antagonizing p110δ constitutes a previously unappreciated therapeutic opportunity for diabetic retinopathy.
Collapse
Affiliation(s)
- Wenyi Wu
- Department of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China
- Schepens Eye Research Institute of Massachusetts Eye and Ear and Department of Ophthalmology, Harvard Medical School, Boston, MA
| | - Guohong Zhou
- Schepens Eye Research Institute of Massachusetts Eye and Ear and Department of Ophthalmology, Harvard Medical School, Boston, MA
- Shanxi Eye Hospital, Taiyuan, China
| | - Haote Han
- Schepens Eye Research Institute of Massachusetts Eye and Ear and Department of Ophthalmology, Harvard Medical School, Boston, MA
| | - Xionggao Huang
- Schepens Eye Research Institute of Massachusetts Eye and Ear and Department of Ophthalmology, Harvard Medical School, Boston, MA
- Department of Ophthalmology, the First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Heng Jiang
- Schepens Eye Research Institute of Massachusetts Eye and Ear and Department of Ophthalmology, Harvard Medical School, Boston, MA
- Department of Ophthalmology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Shizuo Mukai
- Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA
| | - Andrius Kazlauskas
- Department of Ophthalmology and Visual Sciences and Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL
| | - Jing Cui
- The University of British Columbia, Vancouver, British Columbia, Canada
| | | | | | - Xiaobo Xia
- Department of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China
| | - Jiantao Wang
- Shenzhen Eye Hospital, Shenzhen Eye Institute, Shenzhen, China
| | - Hetian Lei
- Shenzhen Eye Hospital, Shenzhen Eye Institute, Shenzhen, China
| |
Collapse
|
12
|
Rusiecki R, Witkowski J, Jaszczewska-Adamczak J. MDM2-p53 Interaction Inhibitors: The Current State-of-Art and Updated Patent Review (2010-Present). Recent Pat Anticancer Drug Discov 2020; 14:324-369. [DOI: 10.2174/1574892814666191022163540] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Revised: 10/09/2019] [Accepted: 10/15/2019] [Indexed: 01/10/2023]
Abstract
Background:
Mouse Double Minute 2 protein (MDM2) is a cellular regulator of p53 tumor
suppressor (p53). Inhibition of the interaction between MDM2 and p53 proteins is a promising anticancer
therapy.
Objective:
This updated patent review is an attempt to compile the research and achievements of the
various researchers working on small molecule MDM2 inhibitors from 2010 to date. We provide an
outlook into the future for therapy based on MDM2 inhibition by presenting an overview of the most
relevant patents which have recently appeared in the literature.
Methods:
Literature and recent patents focusing on the anticancer potential of MDM2-p53 interaction
inhibitors and its applications have been analyzed. We put the main emphasis on the most perspective
compounds which are or were examined in clinical trials.
Results:
Literature data indicated that MDM2 inhibitors are therapeutically effective in specific types
of cancer or non-cancer diseases. A great number of patents and research work around new MDM2-
p53 interaction inhibitors, possible combinations, new indications, clinical regimens in previous years
prove that this targeted therapy is in the scope of interest for many business and academic research
groups.
Conclusion:
Novel MDM2 inhibitors thanks to higher potency and better ADME properties have
shown effectiveness in preclinical and clinical development however the final improvement of therapeutic
potential for MDM2 inhibitors might depend on the useful combination therapy and exploring
new cancer and non-cancer indications.
Collapse
Affiliation(s)
- Rafał Rusiecki
- Faculty of Chemistry, Warsaw University of Technology, Noakowskiego 3, Warsaw 00-664, Poland
| | - Jakub Witkowski
- Faculty of Chemistry, University of Warsaw, Pasteura 1, Warsaw 02-093, Poland
| | | |
Collapse
|
13
|
Xin T, Han H, Wu W, Huang X, Cui J, Matsubara JA, Song J, Wang F, Colyer M, Lei H. Idelalisib inhibits vitreous-induced Akt activation and proliferation of retinal pigment epithelial cells from epiretinal membranes. Exp Eye Res 2019; 190:107884. [PMID: 31786159 DOI: 10.1016/j.exer.2019.107884] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 09/19/2019] [Accepted: 11/19/2019] [Indexed: 01/01/2023]
Abstract
Proliferative vitreoretinopathy (PVR) is a blinding fibrotic eye disease that develops in 8-10% of patients who undergo primary retinal detachment-reparative surgery and in 40-60% of patients with open-globe injury. At present, there is no pharmacological treatment for this devastating disease. Vitreal growth factors activate their respective receptors of cells in the vitreous, trigger their downstream signaling transduction (e.g. phosphoinositide 3 kinases (PI3Ks)/Akt), and drive cellular responses intrinsic to the pathogenesis of PVR. PI3Ks play a central role in experimental PVR. However, which isoform(s) are involved in PVR pathogenesis remain unknown. Herein, we show that p110δ, a catalytic subunit of receptor-regulated PI3K isoform δ, is highly expressed in epiretinal membranes from patients with PVR, and that idelalisib, a specific inhibitor of PI3Kδ, effectively inhibits vitreous-induced Akt activation, proliferation, migration and contraction of retinal pigment epithelial cells derived from an epiretinal membrane of a PVR patient. Small molecules of kinase inhibitors have shown great promise as a class of therapeutics for a variety of human diseases. The data herein suggest that idelalisib is a promising PVR prophylactic.
Collapse
Affiliation(s)
- Tianyi Xin
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA, USA; Department of Ophthalmology, Harvard Medical School, Boston, USA; Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China
| | - Haote Han
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA, USA; Department of Ophthalmology, Harvard Medical School, Boston, USA; College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou, Zhejiang Province, PR China
| | - Wenyi Wu
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA, USA; Department of Ophthalmology, Harvard Medical School, Boston, USA; Department of Ophthalmology, Xiangya Hospital, Central South University, Changsha, Hunan Province, PR China
| | - Xionggao Huang
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA, USA; Department of Ophthalmology, Harvard Medical School, Boston, USA; Department of Ophthalmology, Hainan Medical University, Haikou, Hainan Province, PR China
| | - Jing Cui
- The University of British Columbia, Canada
| | | | - Jingyuan Song
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China
| | - Fang Wang
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Marcus Colyer
- Department of Surgery, Walter Reed-Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Hetian Lei
- Shenzhen Eye Hospital, Shenzhen, Guangdong Province, PR China; Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA, USA; Department of Ophthalmology, Harvard Medical School, Boston, USA.
| |
Collapse
|
14
|
Liu B, Song J, Han H, Hu Z, Chen N, Cui J, Matsubara JA, Zhong J, Lei H. Blockade of MDM2 with inactive Cas9 prevents epithelial to mesenchymal transition in retinal pigment epithelial cells. J Transl Med 2019; 99:1874-1886. [PMID: 31439892 DOI: 10.1038/s41374-019-0307-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 07/08/2019] [Accepted: 07/10/2019] [Indexed: 01/10/2023] Open
Abstract
Epithelial to mesenchymal transition (EMT) plays an important role in the pathogenesis of proliferative vitreoretinopathy (PVR). We aimed to demonstrate the role of mouse double minute 2 (MDM2) in transforming growth factor-beta 2 (TGF-β2)-induced EMT in human retinal pigment epithelial cells (RPEs). Immunofluorescence was used to assess MDM2 expression in epiretinal membranes (ERMs) from patients with PVR. A single guide (sg)RNA targeting the second promoter of MDM2 was cloned into a mutant lentiviral Clustered Regularly Interspaced Short Palindromic Repeats (lentiCRISPR) v2 (D10A and H840A) vector for expressing nuclease dead Cas9 (dCas9)/MDM2-sgRNA in RPEs. In addition, MDM2-sgRNA was also cloned into a pLV-sgRNA-dCas9-Kruppel associated box (KRAB) vector for expressing dCas9 fused with a transcriptional repressor KRAB/MDM2-sgRNA. TGF-β2-induced expression of MDM2 and EMT biomarkers were assessed by quantitative polymerase chain reaction (q-PCR), western blot, or immunofluorescence. Wound-healing and proliferation assays were used to evaluate the role of MDM2 in TGF-β2-induced responses in RPEs. As a result, we found that MDM2 was expressed obviously in ERMs, and that TGF-β2-induced expression of MDM2 and EMT biomarkers Fibronectin, N-cadherin and Vimentin in RPEs. Importantly, we discovered that the dCas9/MDM2-sgRNA blocked TGF-β2-induced expression of MDM2 and the EMT biomarkers without affecting their basal expression, whereas the dCas9-KRAB/MDM2-sgRNA suppressed basal MDM2 expression in RPEs. These cells could not be maintained continuously because their viability was greatly reduced. Next, we found that Nutlin-3, a small molecule blocking the interaction of MDM2 with p53, inhibited TGF-β2-induced expression of Fibronectin and N-cadherin but not Vimentin in RPEs, indicating that MDM2 functions in both p53-dependent and -independent pathways. Finally, our experimental data demonstrated that dCas9/MDM2-sgRNA suppressed TGF-β2-dependent cell proliferation and migration without disturbing the unstimulated basal activity. In conclusion, the CRISPR/dCas9 capability for blocking TGF-β2-induced expression of MDM2 and EMT biomarkers can be exploited for a therapeutic approach to PVR.
Collapse
Affiliation(s)
- Bing Liu
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA.,Department of Ophthalmology, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, China
| | - Jingyuan Song
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA.,Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, 100193, Beijing, China
| | - Haote Han
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA.,Department of Biomedical Engineering, Zhejiang University, 310027, Hangzhou, China
| | - Zhengping Hu
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA
| | - Na Chen
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA.,Department of Ophthalmology, Renji Hospital School of Medicine, Shanghai Jiaotong University, 200127, Shanghai, China
| | - Jing Cui
- The University of British Columbia, Vancouver, BC, V5Z 3N9, Canada
| | | | - Jingxiang Zhong
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, China
| | - Hetian Lei
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA.
| |
Collapse
|
15
|
Chen N, Hu Z, Yang Y, Han H, Lei H. Inactive Cas9 blocks vitreous-induced expression of Mdm2 and proliferation and survival of retinal pigment epithelial cells. Exp Eye Res 2019; 186:107716. [PMID: 31278903 DOI: 10.1016/j.exer.2019.107716] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 06/21/2019] [Accepted: 06/28/2019] [Indexed: 01/09/2023]
Abstract
Mouse double minute (MDM)2 single nucleotide polymorphism (SNP) 309G allele in the second promoter of MDM2 enhances vitreous-induced expression of Mdm2 and degradation of the tumor suppressor protein p53. This MDM2SNP309G contributes to certain cancer development and experimental proliferative vitreoretinopathy. The goal of this study is to discover a novel strategy to only block vitreous-induced expression of Mdm2 for preventing vitreous-induced cell proliferation and survival and thus find a potential novel strategy to treat proliferation-related diseases. We created two mutations (D10A and H840A) in Streptococcus pyogenes (Sp)Cas9 within the nuclease domains (RuvC1 and HNH, respectively) to render this SpCas9 nuclease dead named as dCas9 in a lentiCRISPR v2 vector. Then an MDM2-sgRNA targeting the second promoter of human MDM2 gene was cloned into this vector for producing lentivirus to infect human retinal pigment epithelial (RPE) cells with, which carry a heterozygous genotype of MDM2SNP309 T/G. lacZ-sgRNA was used as a control. As a result, we discovered that vitreous from experimental rabbits induced a 1.9 ± 0.2 fold increase in Mdm2 and a 2.0 ± 0.2 fold decrease in p53 in the RPE cells with dCas9/lacZ-sgRNA compared to those with dCas9/MDM2-sgRNA, suggesting that dCas9 under the guidance of the MDM2-sgRNA prevented RV-stimulated increase in Mdm2. In addition, we found that the rabbit vitreous significantly enhanced cell proliferation (1.5 ± 0.2 fold), survival against apoptosis (2.2 ± 0.2 fold), migration (10 ± 1.5%) and contraction (112.7 ± 14.1 mm2) of the cells with dCas9/lacZ-sgRNA compared with those with dCas9/MDM2-sgRNA. These results indicated that application of the dCas9 targeted to the P2 of MDM2 is a potential therapeutic approach to diseases due to the P2-driven aberrant expression of Mdm2 - such as proliferative vitreoretinopathy.
Collapse
Affiliation(s)
- Na Chen
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; Department of Ophthalmology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, PR China
| | - Zhengping Hu
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Yanhui Yang
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Haote Han
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Hetian Lei
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
16
|
Han H, Chen N, Huang X, Liu B, Tian J, Lei H. Phosphoinositide 3-kinase δ inactivation prevents vitreous-induced activation of AKT/MDM2/p53 and migration of retinal pigment epithelial cells. J Biol Chem 2019; 294:15408-15417. [PMID: 31467081 DOI: 10.1074/jbc.ra119.010130] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 08/22/2019] [Indexed: 12/15/2022] Open
Abstract
Phosphoinositide 3-kinases (PI3Ks) are a family of lipid kinases that play a critical role in transmitting signals from cell-surface molecules to intracellular protein effectors. Key PI3Ks include PI3Kα, PI3Kβ, and PI3Kδ, which are regulated by receptors. The signaling pathway comprising the PI3Ks, along with a Ser/Thr kinase (AKT), a proto-oncogene product (mouse double minute (MDM)2), and a tumor suppressor protein (p53), plays an essential role in experimental proliferative vitreoretinopathy (PVR), which is a fibrotic blinding eye disorder. However, which PI3K isoforms are involved in PVR is unknown. A major characteristic of PVR is the formation of epi (or sub)-retinal membranes that consist of extracellular matrix and cells, including retinal pigment epithelium (RPE) cells, glial cells, and macrophages. RPE cells are considered key players in PVR pathogenesis. Using immunoblotting and immunofluorescence analyses, we herein provide the evidence that PI3Kδ is highly expressed in human RPEs when it is primarily expressed in leukocytes. We also found that PI3Kδ inactivation through two approaches, CRISPR/Cas9-mediated depletion and a PI3Kδ-specific inhibitor (idelalisib), not only blocks vitreous-induced activation of AKT and MDM2 but also abrogates a vitreous-stimulated decrease in p53. Furthermore, we demonstrate that PI3Kδ inactivation prevents vitreous-induced proliferation, migration, and contraction of human RPEs. These results suggest that PI3Kδ may represent a potential therapeutic target for RPE-related eye diseases, including PVR.
Collapse
Affiliation(s)
- Haote Han
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, Massachusetts 02114.,Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts 02115.,College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou 310027, China.,Zhejiang-Malaysia Joint Research Center for Traditional Medicine, Zhejiang University, Hangzhou 310027 China
| | - Na Chen
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, Massachusetts 02114.,Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts 02115.,Department of Ophthalmology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Xionggao Huang
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, Massachusetts 02114.,Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts 02115.,Department of Ophthalmology, The First Affiliated Hospital of Hainan Medical University, Haikou 570102, China
| | - Bing Liu
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, Massachusetts 02114.,Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts 02115
| | - Jingkui Tian
- College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou 310027, China.,Zhejiang-Malaysia Joint Research Center for Traditional Medicine, Zhejiang University, Hangzhou 310027 China
| | - Hetian Lei
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, Massachusetts 02114 .,Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts 02115
| |
Collapse
|
17
|
Zhou G, Duan Y, Ma G, Wu W, Hu Z, Chen N, Chee Y, Cui J, Samad A, Matsubara JA, Mukai S, D'Amore PA, Lei H. Introduction of the MDM2 T309G Mutation in Primary Human Retinal Epithelial Cells Enhances Experimental Proliferative Vitreoretinopathy. Invest Ophthalmol Vis Sci 2017; 58:5361-5367. [PMID: 29049737 PMCID: PMC5649510 DOI: 10.1167/iovs.17-22045] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Purpose The murine double minute (MDM)2 is a critical negative regulator of the p53 tumor suppressor, and MDM2 SNP309G is associated with a higher risk of proliferative vitreoretinopathy (PVR); in addition, the MDM2 T309G created using clustered regularly interspaced short palindromic repeats (CRISPR)/associated endonuclease (Cas)9 enhances normal rabbit vitreous-induced expression of MDM2 and survival of primary human retinal pigment epithelial (hRPE) cells in vitro. The goal of this study was to determine whether this MDM2 T309G contributes to the development of experimental PVR. Methods hRPE cells expressing MDM2 T309G or T309T only were treated with vitreous from human PVR donors (HV). The expression of MDM2 and p53 in the treated cells was examined by Western blot. The in vitro vitreous-induced cellular responses, such as contraction were assessed, and PVR was induced by intravitreal injection of the hRPE cells with MDM2 T309G or T309T only into rabbit eyes. Results Western blot analyses indicated that treatment of hRPE cells with HV led to a significant increase (1.7 ± 0.2-fold) in the expression of MDM2 and a significant decrease in p53 in the cells expressing MDM2 T309G compared with those with MDM2 T309T. In addition, HV promoted contraction of the hRPE cells expressing MDM2 T309G significantly more than those with MDM2 T309T only. Furthermore, MDM2 T309G in the hRPE cells enhanced the development of PVR in a rabbit model. Conclusions The MDM2 SNP309 in RPE cells enhances their potential of PVR pathogenesis.
Collapse
Affiliation(s)
- Guohong Zhou
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, United States.,Shanxi Eye Hospital, Taiyuan City, Shanxi Province, China
| | - Yajiang Duan
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, United States.,Shanxi Eye Hospital, Taiyuan City, Shanxi Province, China
| | - Gaoen Ma
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, United States.,Department of Ophthalmology, the Third Affiliated Hospital of Xinxiang Medical University, Eye Hospital of Xinxiang Medical University, Xinxiang, Henan Province, China
| | - Wenyi Wu
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, United States
| | - Zhengping Hu
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, United States
| | - Na Chen
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, United States
| | - Yewlin Chee
- Massachusetts Eye and Ear Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States
| | - Jing Cui
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Arif Samad
- Dalhousie University, Halifax, Nova Scotia, Canada
| | - Joanne A Matsubara
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Shizuo Mukai
- Massachusetts Eye and Ear Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States
| | - Patricia A D'Amore
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, United States
| | - Hetian Lei
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, United States
| |
Collapse
|
18
|
Wang HF, Ma JX, Shang QL, An JB, Chen HT. Crocetin inhibits the proliferation, migration and TGF-β 2-induced epithelial-mesenchymal transition of retinal pigment epithelial cells. Eur J Pharmacol 2017; 815:391-398. [PMID: 28970011 DOI: 10.1016/j.ejphar.2017.09.041] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 09/20/2017] [Accepted: 09/28/2017] [Indexed: 10/18/2022]
Abstract
Retinal pigment epithelial (RPE) cells, the major cell type in the fibrotic membrane of proliferative vitreoretinopathy, display enhanced proliferative and migratory capacities and epithelial-mesenchymal transition (EMT). In this study, we investigated the potential impact of crocetin on the proliferation, migration and EMT of cultured ARPE-19 cells. The cells were treated with crocetin alone or in combination with transforming growth factor-β2 (TGF-β2). Cell proliferation was examined using the CCK-8 assay. Cell cycle distribution was analyzed by flow cytometry after propidium iodide staining. The expression levels of proliferating cell nuclear antigen (PCNA), p21 and p53 were examined by Western blot analysis. Cell migration was assessed by in vitro scratch and Transwell assays. Real-time PCR, Western blotting and immunofluorescence were used to assess EMT features. Treatment of ARPE-19 cells with crocetin (50-200μM) significantly inhibited their proliferation and migration in a concentration- and time-dependent manner. Crocetin induced G1 arrest, reduced PCNA protein expression and increased the p21 and p53 accumulation in ARPE-19 cells. Crocetin inhibited TGF-β2-induced EMT in ARPE-19 cells by maintaining the expression of E-cadherin and ZO-1 and by reducing the expression of vimentin and α-SMA through the suppression of phosphorylation of p38. These results indicate that crocetin is an effective inhibitor of the proliferation, migration and TGF-β2-mediated EMT of ARPE-19 cells.
Collapse
Affiliation(s)
- Hui-Fang Wang
- Department of Ophthalmology, The Second Hospital of HeBei Medical University, Shijiazhuang 050000, China; Department of Ophthalmology, The First Hospital of Shijiazhuag City, Shijiazhuang 050000, China
| | - Jing-Xue Ma
- Department of Ophthalmology, The Second Hospital of HeBei Medical University, Shijiazhuang 050000, China.
| | - Qing-Li Shang
- Department of Ophthalmology, The Second Hospital of HeBei Medical University, Shijiazhuang 050000, China
| | - Jian-Bin An
- Department of Ophthalmology, The Second Hospital of HeBei Medical University, Shijiazhuang 050000, China
| | - Hai-Ting Chen
- Department of Ophthalmology, The Second Hospital of HeBei Medical University, Shijiazhuang 050000, China; Department of Ophthalmology, Cangzhou Central Hospital, Cangzhou 061000, China
| |
Collapse
|
19
|
Ma G, Duan Y, Huang X, Qian CX, Chee Y, Mukai S, Cui J, Samad A, Matsubara JA, Kazlauskas A, D'Amore PA, Gu S, Lei H. Prevention of Proliferative Vitreoretinopathy by Suppression of Phosphatidylinositol 5-Phosphate 4-Kinases. Invest Ophthalmol Vis Sci 2017; 57:3935-43. [PMID: 27472081 PMCID: PMC4974024 DOI: 10.1167/iovs.16-19405] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Purpose Previous studies have shown that vitreous stimulates degradation of the tumor suppressor protein p53 and that knockdown of phosphatidylinositol 5-phosphate 4-kinases (PI5P4Kα and -β) abrogates proliferation of p53-deficient cells. The purpose of this study was to determine whether vitreous stimulated expression of PI5P4Kα and -β and whether suppression of PI5P4Kα and -β would inhibit vitreous-induced cellular responses and experimental proliferative vitreoretinopathy (PVR). Methods PI5P4Kα and -β encoded by PIP4K2A and 2B, respectively, in human ARPE-19 cells were knocked down by stably expressing short hairpin (sh)RNA directed at human PIP4K2A and -2B. In addition, we rescued expression of PI5P4Kα and -β by re-expressing mouse PIP4K2A and -2B in the PI5P4Kα and -β knocked-down ARPE-19 cells. Expression of PI5P4Kα and -β was determined by Western blot and immunofluorescence. The following cellular responses were monitored: cell proliferation, survival, migration, and contraction. Moreover, the cell potential of inducing PVR was examined in a rabbit model of PVR effected by intravitreal cell injection. Results We found that vitreous enhanced expression of PI5P4Kα and -β in RPE cells and that knocking down PI5P4Kα and -β abrogated vitreous-stimulated cell proliferation, survival, migration, and contraction. Re-expression of mouse PIP4Kα and -β in the human PI5P4Kα and -β knocked-down cells recovered the loss of vitreous-induced cell contraction. Importantly, suppression of PI5P4Kα and -β abrogated the pathogenesis of PVR induced by intravitreal cell injection in rabbits. Moreover, we revealed that expression of PI5P4Kα and -β was abundant in epiretinal membranes from PVR grade C patients. Conclusions The findings from this study indicate that PI5P4Kα and -β could be novel therapeutic targets for the treatment of PVR.
Collapse
Affiliation(s)
- Gaoen Ma
- Schepens Eye Research Institute, Boston, Massachusetts, United States 2Massachusetts Eye and Ear, Boston, Massachusetts, United States 3Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States 4Aier School of Ophthalmology
| | - Yajian Duan
- Schepens Eye Research Institute, Boston, Massachusetts, United States 2Massachusetts Eye and Ear, Boston, Massachusetts, United States 3Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States
| | - Xionggao Huang
- Schepens Eye Research Institute, Boston, Massachusetts, United States 3Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States
| | - Cynthia X Qian
- Massachusetts Eye and Ear, Boston, Massachusetts, United States 3Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States
| | - Yewlin Chee
- Massachusetts Eye and Ear, Boston, Massachusetts, United States 3Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States
| | - Shizuo Mukai
- Massachusetts Eye and Ear, Boston, Massachusetts, United States 3Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States
| | - Jing Cui
- The University of British Columbia, Vancouver, British Columbia, Canada
| | - Arif Samad
- Dalhousie University, Halifax, Nova Scotia, Canada
| | | | - Andrius Kazlauskas
- Schepens Eye Research Institute, Boston, Massachusetts, United States 3Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States
| | - Patricia A D'Amore
- Schepens Eye Research Institute, Boston, Massachusetts, United States 3Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States
| | - Shuyan Gu
- Aier School of Ophthalmology, Central South University, Changsha, Changsha Province, China
| | - Hetian Lei
- Schepens Eye Research Institute, Boston, Massachusetts, United States 3Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States
| |
Collapse
|
20
|
MicroRNA-182 Suppresses HGF/SF-Induced Increases in Retinal Pigment Epithelial Cell Proliferation and Migration through Targeting c-Met. PLoS One 2016; 11:e0167684. [PMID: 27936052 PMCID: PMC5147940 DOI: 10.1371/journal.pone.0167684] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 11/18/2016] [Indexed: 01/29/2023] Open
Abstract
As increases in hepatocyte growth factor/scatter factor (HGF/SF) induce retinal pigment epithelial (RPE) migration and proliferation into the vitreous cavity and contribute to proliferative vitreoretinopathy (PVR) development, we determined if changes in miR-182 expression affect such behavioral changes. We found that miR-182 expression was less in PVR clinical samples than in primary RPE cells whereas c-Met was upregulated. Ectopic miR-182 inhibited RPE cell proliferation, cell cycle, and migration. Bioinformatic analysis identified c-Met as a miR-182 target, which was confirmed with the luciferase reporter assay. Transfection of miR-182 into RPE cells induced c-Met downregulation, which led to reduced cell proliferation and migration through declines in p-Akt formation. MiR-182 downregulation along with c-Met upregulation in PVR tissues suggest that these two opposing effects play important roles in PVR development. As ectopic miR-182 expression suppressed RPE cell proliferation and migration, strategies to selectively upregulate miR-182 expression in a clinical setting may provide a novel option to treat this disease.
Collapse
|
21
|
Zhang J, Yuan G, Dong M, Zhang T, Hua G, Zhou Q, Shi W. Notch signaling modulates proliferative vitreoretinopathy via regulating retinal pigment epithelial-to-mesenchymal transition. Histochem Cell Biol 2016; 147:367-375. [PMID: 27600720 DOI: 10.1007/s00418-016-1484-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/23/2016] [Indexed: 12/25/2022]
Abstract
Elevated Notch signaling has been verified in a large range of fibrotic diseases developed in the kidney, liver, and lung, inducing the development of the epithelial-mesenchymal transition (EMT). The aim of this study was to observe the involvement of Notch signaling in the EMT of retinal pigment epithelial (RPE) cells and the pathogenesis of proliferative vitreoretinopathy (PVR). In vitro cultivated human RPE cells (ARPE-19) were treated with 10 ng/mL transforming growth factor (TGF)-β1 for 24, 48, and 72 h. The expression levels of ZO-1, α-SMA, vimentin, Notch1 intracellular domain (NICD1), and Hes-1 were evaluated with quantitative real-time polymerase chain reaction (qRT-PCR), immunofluorescence staining or Western blot. TGF-β1 induced EMT and the activation of Notch signaling in ARPE-19 cells. To examine the effect of Notch inhibition on TGF-β1-induced EMT and PVR formation, ARPE-19 cells were preincubated with γ-secretase inhibitor LY411575 before TGF-β1 treatment. Mouse PVR model was used for in vivo study. ARPE-19 cells were injected intravitreously with or without the LY411575 to examine the effect of Notch inhibition on PVR formation. LY411575 significantly attenuated EMT by inhibiting the Notch signaling activation in vitro. PVR was induced by intravitreal injections of ARPE-19 cells, while LY411575 inhibited mouse PVR formation in vivo. Notch signaling plays a critical role in TGF-β1-induced EMT in vitro and mice PVR model, which provides a novel insight into the pathogenesis of PVR. The specific inhibition of Notch signaling by γ-secretase inhibitor may provide a new approach for the prevention of PVR.
Collapse
Affiliation(s)
- Jingjing Zhang
- Shandong Eye Hospital, Shandong Eye Institute, Shandong Academy of Medical Sciences, 372 Jingsi Road, Jinan, 250021, China
| | - Gongqiang Yuan
- Shandong Eye Hospital, Shandong Eye Institute, Shandong Academy of Medical Sciences, 372 Jingsi Road, Jinan, 250021, China
| | - Muchen Dong
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong Academy of Medical Sciences, Qingdao, 266071, Shandong Province, China
| | - Ting Zhang
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong Academy of Medical Sciences, Qingdao, 266071, Shandong Province, China
| | - Gao Hua
- Shandong Eye Hospital, Shandong Eye Institute, Shandong Academy of Medical Sciences, 372 Jingsi Road, Jinan, 250021, China
| | - Qingjun Zhou
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong Academy of Medical Sciences, Qingdao, 266071, Shandong Province, China.
| | - Weiyun Shi
- Shandong Eye Hospital, Shandong Eye Institute, Shandong Academy of Medical Sciences, 372 Jingsi Road, Jinan, 250021, China.
| |
Collapse
|
22
|
Duan Y, Ma G, Huang X, D'Amore PA, Zhang F, Lei H. The Clustered, Regularly Interspaced, Short Palindromic Repeats-associated Endonuclease 9 (CRISPR/Cas9)-created MDM2 T309G Mutation Enhances Vitreous-induced Expression of MDM2 and Proliferation and Survival of Cells. J Biol Chem 2016; 291:16339-47. [PMID: 27246850 DOI: 10.1074/jbc.m116.729467] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Indexed: 01/09/2023] Open
Abstract
The G309 allele of SNPs in the mouse double minute (MDM2) promoter locus is associated with a higher risk of cancer and proliferative vitreoretinopathy (PVR), but whether SNP G309 contributes to the pathogenesis of PVR is to date unknown. The clustered regularly interspaced short palindromic repeats (CRISPR)-associated endonuclease (Cas) 9 from Streptococcus pyogenes (SpCas9) can be harnessed to manipulate a single or multiple nucleotides in mammalian cells. Here we delivered SpCas9 and guide RNAs using dual adeno-associated virus-derived vectors to target the MDM2 genomic locus together with a homologous repair template for creating the mutation of MDM2 T309G in human primary retinal pigment epithelial (hPRPE) cells whose genotype is MDM2 T309T. The next-generation sequencing results indicated that there was 42.51% MDM2 G309 in the edited hPRPE cells using adeno-associated viral CRISPR/Cas9. Our data showed that vitreous induced an increase in MDM2 and subsequent attenuation of p53 expression in MDM2 T309G hPRPE cells. Furthermore, our experimental results demonstrated that MDM2 T309G in hPRPE cells enhanced vitreous-induced cell proliferation and survival, suggesting that this SNP contributes to the pathogenesis of PVR.
Collapse
Affiliation(s)
- Yajian Duan
- From the Schepens Eye Research Institute, Massachusetts Eye and Ear, and Departments of Ophthalmology and the Shanxi Eye Hospital, Taiyuan, Shanxi 030000, China
| | - Gaoen Ma
- From the Schepens Eye Research Institute, Massachusetts Eye and Ear, and Departments of Ophthalmology and
| | - Xionggao Huang
- From the Schepens Eye Research Institute, Massachusetts Eye and Ear, and Departments of Ophthalmology and
| | - Patricia A D'Amore
- From the Schepens Eye Research Institute, Massachusetts Eye and Ear, and Departments of Ophthalmology and Pathology, Harvard Medical School, Boston, Massachusetts 02114
| | - Feng Zhang
- the Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts 02142, and
| | - Hetian Lei
- From the Schepens Eye Research Institute, Massachusetts Eye and Ear, and Departments of Ophthalmology and
| |
Collapse
|
23
|
Notch signaling regulates M2 type macrophage polarization during the development of proliferative vitreoretinopathy. Cell Immunol 2015; 298:77-82. [DOI: 10.1016/j.cellimm.2015.09.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 09/02/2015] [Accepted: 09/15/2015] [Indexed: 02/02/2023]
|
24
|
Pastor-Idoate S, Rodríguez-Hernández I, Rojas J, Fernández I, García-Gutierrez MT, Ruiz-Moreno JM, Rocha-Sousa A, Ramkissoon YD, Harsum S, MacLaren RE, Charteris DG, Van Meurs JC, González-Sarmiento R, Pastor JC. BAX and BCL-2 polymorphisms, as predictors of proliferative vitreoretinopathy development in patients suffering retinal detachment: the Retina 4 project. Acta Ophthalmol 2015; 93:e541-9. [PMID: 25991504 DOI: 10.1111/aos.12718] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2014] [Accepted: 02/18/2015] [Indexed: 12/31/2022]
Abstract
PURPOSE To compare the distribution of BCL-2 -938C>A (rs2279115) and BAX -248G>A (rs4645878) genotypes among European subjects undergoing rhegmatogenous retinal detachment (RRD) surgery in relation to the further development of proliferative vitreoretinopathy (PVR). METHODS A case-control gene association study, as a part of Retina 4 project, was designed. rs2279115 and rs4645878 polymorphisms were analysed in 555 samples from patients with RRD (134 with PVR secondary to surgery). Proportions of genotypes and AA homozygous groups of BCL-2 and BAX polymorphisms between subsamples were analysed in two phases. Genotypic and allelic frequencies were compared in global sample and in subsamples. RESULTS BAX: Differences were observed in the genotype frequencies and in AA carriers between controls and cases in the global series. The odds ratio (OR) of A carriers in the global sample was 1.7 (95% CI: 1.23-2.51). Proportions of genotypes in Spain + Portugal were significant different. The OR of A carriers from Spain and Portugal was 1.8 (95% CI: 1.11-2.95). BCL-2: No significant differences were observed in genotype frequencies. However, proportions of genotypes in Spain + Portugal were significant. A protective effect (OR: 0.6 95% CI: 0.43-0.96) was found in A carriers from Spain and Portugal. CONCLUSIONS Results suggest that A allele of rs4645878 could be a biomarker of high risk of developing PVR in patients undergoing RD surgery. The possible role of BCL-2 (inhibitor of necroptosis pathway) as a possible new target in PVR prophylaxis should be investigated.
Collapse
Affiliation(s)
- Salvador Pastor-Idoate
- Instituto de Oftalmobiología Aplicada (IOBA-Retina Group); University of Valladolid; Valladolid Spain
- Unidad de Medicina Molecular; Departamento de Medicina; University of Salamanca; Salamanca Spain
| | - Irene Rodríguez-Hernández
- Unidad de Medicina Molecular; Departamento de Medicina; University of Salamanca; Salamanca Spain
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC); Consejo Superior de Investigaciones Científicas (CSIC); Instituto de Investigación Biomédica de Salamanca (IBSAL); University of Salamanca; Salamanca Spain
| | - Jimena Rojas
- Instituto de Oftalmobiología Aplicada (IOBA-Retina Group); University of Valladolid; Valladolid Spain
| | - Itziar Fernández
- Instituto de Oftalmobiología Aplicada (IOBA-Retina Group); University of Valladolid; Valladolid Spain
| | | | | | - Amandio Rocha-Sousa
- Department of Sense Organs; Medical School; Hospital SanJoão; University of Porto; Porto Portugal
| | - Yashin D. Ramkissoon
- Moorfields Eye Hospital; National Institute of Health Research (NIHR); Biomedical Research Centre; London UK
- Royal Hallamshire Hospital; University of Sheffield; Sheffield UK
| | - Steven Harsum
- Moorfields Eye Hospital; National Institute of Health Research (NIHR); Biomedical Research Centre; London UK
| | - Robert E. MacLaren
- Moorfields Eye Hospital; National Institute of Health Research (NIHR); Biomedical Research Centre; London UK
- Nuffield Laboratory of Ophthalmology; John Radcliffe Hospital; University of Oxford; Oxford UK
| | - David G. Charteris
- Moorfields Eye Hospital; National Institute of Health Research (NIHR); Biomedical Research Centre; London UK
| | - Jan C. Van Meurs
- Rotterdam Eye Hospital; Erasmus Medical Center; University of Rotterdam; Rotterdam The Netherlands
| | - Rogelio González-Sarmiento
- Unidad de Medicina Molecular; Departamento de Medicina; University of Salamanca; Salamanca Spain
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC); Consejo Superior de Investigaciones Científicas (CSIC); Instituto de Investigación Biomédica de Salamanca (IBSAL); University of Salamanca; Salamanca Spain
| | - Jose C. Pastor
- Instituto de Oftalmobiología Aplicada (IOBA-Retina Group); University of Valladolid; Valladolid Spain
| | | |
Collapse
|
25
|
Kon Graversen V, Chavala SH. MDM2 inhibitors in the search for an optimized neovascular age-related macular degeneration treatment. EXPERT REVIEW OF OPHTHALMOLOGY 2014. [DOI: 10.1586/17469899.2014.952225] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
26
|
Pennock S, Haddock LJ, Mukai S, Kazlauskas A. Vascular endothelial growth factor acts primarily via platelet-derived growth factor receptor α to promote proliferative vitreoretinopathy. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:3052-68. [PMID: 25261788 DOI: 10.1016/j.ajpath.2014.07.026] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2014] [Revised: 07/09/2014] [Accepted: 07/24/2014] [Indexed: 01/22/2023]
Abstract
Proliferative vitreoretinopathy (PVR) is a nonneovascular blinding disease and the leading cause for failure in surgical repair of rhegmatogenous retinal detachments. Once formed, PVR is difficult to treat. Hence, there is an acute interest in developing approaches to prevent PVR. Of the many growth factors and cytokines that accumulate in vitreous as PVR develops, neutralizing vascular endothelial growth factor (VEGF) A has recently been found to prevent PVR in at least one animal model. The goal of this study was to test if Food and Drug Administration-approved agents could protect the eye from PVR in multiple animal models and to further investigate the underlying mechanisms. Neutralizing VEGF with aflibercept (VEGF Trap-Eye) safely and effectively protected rabbits from PVR in multiple models of disease. Furthermore, aflibercept reduced the bioactivity of both experimental and clinical PVR vitreous. Finally, although VEGF could promote some PVR-associated cellular responses via VEGF receptors expressed on the retinal pigment epithelial cells that drive this disease, VEGF's major contribution to vitreal bioactivity occurred via platelet-derived growth factor receptor α. Thus, VEGF promotes PVR by a noncanonical ability to engage platelet-derived growth factor receptor α. These findings indicate that VEGF contributes to nonangiogenic diseases and that anti-VEGF-based therapies may be effective on a wider spectrum of diseases than previously appreciated.
Collapse
Affiliation(s)
- Steven Pennock
- The Schepens Eye Research Institute and Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, Massachusetts; Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Luis J Haddock
- The Schepens Eye Research Institute and Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, Massachusetts; Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Shizuo Mukai
- The Schepens Eye Research Institute and Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, Massachusetts; Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Andrius Kazlauskas
- The Schepens Eye Research Institute and Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, Massachusetts; Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
27
|
Pennock S, Haddock LJ, Eliott D, Mukai S, Kazlauskas A. Is neutralizing vitreal growth factors a viable strategy to prevent proliferative vitreoretinopathy? Prog Retin Eye Res 2014; 40:16-34. [PMID: 24412519 DOI: 10.1016/j.preteyeres.2013.12.006] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Revised: 12/23/2013] [Accepted: 12/27/2013] [Indexed: 11/18/2022]
Abstract
Proliferative vitreoretinopathy (PVR) is a blinding disorder that occurs in eyes with rhegmatogenous retinal detachment and in eyes that have recently undergone retinal detachment surgery. There are presently no treatment strategies to reduce the risk of developing PVR in eyes with retinal detachment, and surgical intervention is the only option for eyes with retinal detachment and established PVR. Given the poor visual outcome associated with the surgical treatment of PVR, considerable work has been done to identify pharmacologic agents that could antagonize the PVR process. Intensive efforts to identify molecular determinants of PVR implicate vitreal growth factors. A surprise that emerged in the course of testing the 'growth factor hypothesis' of PVR was the existence of a functional relationship amongst growth factors that engage platelet-derived growth factor (PDGF) receptor α (PDGFRα), a receptor tyrosine kinase that is key to pathogenesis of experimental PVR. Vascular endothelial cell growth factor A (VEGF), which is best known for its ability to activate VEGF receptors (VEGFRs) and induce permeability and/or angiogenesis, enables activation of PDGFRα by a wide spectrum of vitreal growth factors outside of the PDGF family (non-PDGFs) in a way that triggers signaling events that potently enhance the viability of cells displaced into vitreous. Targeting these growth factors or signaling events effectively neutralizes the bioactivity of PVR vitreous and prevents PVR in a number of preclinical models. In this review, we discuss recent conceptual advances in understanding the role of growth factors in PVR, and consider the tangible treatment strategies for clinical application.
Collapse
Affiliation(s)
- Steven Pennock
- The Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
| | - Luis J Haddock
- The Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
| | - Dean Eliott
- The Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
| | - Shizuo Mukai
- The Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
| | - Andrius Kazlauskas
- The Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
28
|
Pastor-Idoate S, Rodríguez-Hernández I, Rojas J, Fernández I, García-Gutiérrez MT, Ruiz-Moreno JM, Rocha-Sousa A, Ramkissoon Y, Harsum S, MacLaren RE, Charteris D, VanMeurs JC, González-Sarmiento R, Pastor JC, on behalf of the Genetics on PVR Study Group. The T309G MDM2 gene polymorphism is a novel risk factor for proliferative vitreoretinopathy. PLoS One 2013; 8:e82283. [PMID: 24349246 PMCID: PMC3857251 DOI: 10.1371/journal.pone.0082283] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Accepted: 10/31/2013] [Indexed: 11/25/2022] Open
Abstract
Proliferative vitreoretinopathy (PVR) is still the major cause of failure in retinal detachment (RD) surgery. It is believed that down-regulation in the p53 pathway could be an important key in PVR pathogenesis. The purpose was to evaluate the impact of T309G MDM2 polymorphism (rs2279744) in PVR. Distribution of T309G MDM2 genotypes among European subjects undergoing RD surgery was evaluated. Proportions of genotypes between subsamples from different countries were analyzed. Also, a genetic interaction between rs2279744 in MDM2 and rs1042522 in p53 gene was analyzed. Significant differences were observed comparing MDM2 genotype frequencies at position 309 of intron 1 between cases (GG: 21.6%, TG: 54.5%, TT: 23.8%) and controls (GG: 7.3%, TG: 43.9%, TT: 48.7%). The proportions of genotypes between sub-samples from different countries showed a significant difference. Distribution of GG genotype revealed differences in Spain (35.1–53.0)/(22.6–32.9), Portugal (39.0–74.4)/(21.4–38.9), Netherlands (40.6–66.3)/(25.3–38.8) and UK (37.5–62.4)/(23.3–34.2). The OR of G carriers in the global sample was 5.9 (95% CI: 3.2 to 11.2). The OR of G carriers from Spain and Portugal was 5.4 (95% CI: 2.2–12.7), whereas in the UK and the Netherlands was 7.3 (95% CI: 2.8–19.1). Results indicate that the G allele of rs2279744 is associated with a higher risk of developing PVR in patients undergoing a RD surgery. Further studies are necessary to understand the role of this SNP in the development of PVR.
Collapse
Affiliation(s)
- Salvador Pastor-Idoate
- Instituto de Oftalmobiología (IOBA-Retina Group), University of Valladolid, Valladolid, Spain
- Unidad de Medicina Molecular, Departamento de Medicina, University of Salamanca, Salamanca, Spain
| | - Irene Rodríguez-Hernández
- Unidad de Medicina Molecular, Departamento de Medicina, University of Salamanca, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL) and Instituto de Biología Molecular y Celular del Cáncer (IBMCC), University of Salamanca–CSIC-SACYL, Salamanca, Spain
| | - Jimena Rojas
- Instituto de Oftalmobiología (IOBA-Retina Group), University of Valladolid, Valladolid, Spain
| | - Itziar Fernández
- Instituto de Oftalmobiología (IOBA-Retina Group), University of Valladolid, Valladolid, Spain
| | | | | | - Amandio Rocha-Sousa
- Department of Sense Organs, Medical School, University of Porto, Hospital San João, Porto, Portugal
| | - Yashin Ramkissoon
- Moorfields Eye Hospital, National Institute of Health Research (NIHR), Biomedical Research Centre, London, United Kingdom
| | - Steven Harsum
- Moorfields Eye Hospital, National Institute of Health Research (NIHR), Biomedical Research Centre, London, United Kingdom
| | - Robert E. MacLaren
- Moorfields Eye Hospital, National Institute of Health Research (NIHR), Biomedical Research Centre, London, United Kingdom
- Nuffield Laboratory of Ophthalmology, University of Oxford, John Radcliffe Hospital Oxford, United Kingdom
| | - David Charteris
- Moorfields Eye Hospital, National Institute of Health Research (NIHR), Biomedical Research Centre, London, United Kingdom
| | - Jan C. VanMeurs
- Rotterdam Eye Hospital, Erasmus Medical Center, University of Rotterdam, The Netherlands
| | - Rogelio González-Sarmiento
- Unidad de Medicina Molecular, Departamento de Medicina, University of Salamanca, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL) and Instituto de Biología Molecular y Celular del Cáncer (IBMCC), University of Salamanca–CSIC-SACYL, Salamanca, Spain
- * E-mail: (RGS); (JCP)
| | - José C. Pastor
- Instituto de Oftalmobiología (IOBA-Retina Group), University of Valladolid, Valladolid, Spain
- * E-mail: (RGS); (JCP)
| | | |
Collapse
|
29
|
Sharma M, Tiwari A, Sharma S, Bhoria P, Gupta V, Gupta A, Luthra-Guptasarma M. Fibrotic remodeling of the extracellular matrix through a novel (engineered, dual-function) antibody reactive to a cryptic epitope on the N-terminal 30 kDa fragment of fibronectin. PLoS One 2013; 8:e69343. [PMID: 23935989 PMCID: PMC3720593 DOI: 10.1371/journal.pone.0069343] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2013] [Accepted: 06/07/2013] [Indexed: 11/18/2022] Open
Abstract
Fibrosis is characterized by excessive accumulation of scar tissue as a result of exaggerated deposition of extracellular matrix (ECM), leading to tissue contraction and impaired function of the organ. Fibronectin (Fn) is an essential component of the ECM, and plays an important role in fibrosis. One such fibrotic pathology is that of proliferative vitreoretinopathy (PVR), a sight-threatening complication which develops as a consequence of failure of surgical repair of retinal detachment. Such patients often require repeated surgeries for retinal re-attachment; therefore, a preventive measure for PVR is of utmost importance. The contractile membranes formed in PVR, are composed of various cell types including the retinal pigment epithelial cells (RPE); fibronectin is an important constituent of the ECM surrounding these cells. Together with the vitreous, fibronectin creates microenvironments in which RPE cells proliferate. We have successfully developed a dual-action, fully human, fibronectin-specific single chain variable fragment antibody (scFv) termed Fn52RGDS, which acts in two ways: i) binds to cryptic sites in fibronectin, and thereby prevents its self polymerization/fibrillogenesis, and ii) interacts with the cell surface receptors, ie., integrins (through an attached “RGD” sequence tag), and thereby blocks the downstream cell signaling events. We demonstrate the ability of this antibody to effectively reduce some of the hallmark features of fibrosis - migration, adhesion, fibronectin polymerization, matrix metalloprotease (MMP) expression, as well as reduction of collagen gel contraction (a model of fibrotic tissue remodeling). The data suggests that the antibody can be used as a rational, novel anti-fibrotic candidate.
Collapse
Affiliation(s)
- Maryada Sharma
- Department of Immunopathology,Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Anil Tiwari
- Department of Immunopathology,Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Shweta Sharma
- Department of Immunopathology,Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Preeti Bhoria
- Department of Internal Medicine,Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Vishali Gupta
- Department of Ophthalmology,Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Amod Gupta
- Department of Ophthalmology,Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Manni Luthra-Guptasarma
- Department of Immunopathology,Postgraduate Institute of Medical Education and Research, Chandigarh, India
- * E-mail:
| |
Collapse
|
30
|
Ranibizumab is a potential prophylaxis for proliferative vitreoretinopathy, a nonangiogenic blinding disease. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 182:1659-70. [PMID: 23582767 DOI: 10.1016/j.ajpath.2013.01.052] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Revised: 01/11/2013] [Accepted: 01/14/2013] [Indexed: 11/20/2022]
Abstract
Proliferative vitreoretinopathy (PVR) exemplifies a disease that is difficult to predict, lacks effective treatment options, and substantially reduces the quality of life of an individual. Surgery to correct a rhegmatogenous retinal detachment fails primarily because of PVR. Likely mediators of PVR are growth factors in vitreous, which stimulate cells within and behind the retina as an inevitable consequence of a breached retina. Three classes of growth factors [vascular endothelial growth factor A (VEGF-A), platelet-derived growth factors (PDGFs), and non-PDGFs (growth factors outside of the PDGF family)] are relevant to PVR pathogenesis because they act on PDGF receptor α, which is required for experimental PVR and is associated with this disease in humans. We discovered that ranibizumab (a clinically approved agent that neutralizes VEGF-A) reduced the bioactivity of vitreous from patients and experimental animals with PVR, and protected rabbits from developing disease. The apparent mechanism of ranibizumab action involved derepressing PDGFs, which, at the concentrations present in PVR vitreous, inhibited non-PDGF-mediated activation of PDGF receptor α. These preclinical findings suggest that available approaches to neutralize VEGF-A are prophylactic for PVR, and that anti-VEGF-based therapies may be effective for managing more than angiogenesis- and edema-driven pathological conditions.
Collapse
|