1
|
Muniyandi A, Hartman GD, Sishtla K, Rai R, Gomes C, Day K, Song Y, Masters AR, Quinney SK, Qi X, Woods H, Boulton ME, Meyer JS, Vilseck JZ, Georgiadis MM, Kelley MR, Corson TW. Ref-1 is overexpressed in neovascular eye disease and targetable with a novel inhibitor. Angiogenesis 2025; 28:11. [PMID: 39756006 DOI: 10.1007/s10456-024-09966-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 12/16/2024] [Indexed: 01/07/2025]
Abstract
Reduction-oxidation factor-1 or apurinic/apyrimidinic endonuclease 1 (Ref-1/APE1) is a crucial redox-sensitive activator of transcription factors such as NF-κB, HIF-1α, STAT-3 and others. It could contribute to key features of ocular neovascularization including inflammation and angiogenesis; these underlie diseases like neovascular age-related macular degeneration (nAMD). We previously revealed a role for Ref-1 in the growth of ocular endothelial cells and in choroidal neovascularization (CNV). Here, we set out to further explore Ref-1 in neovascular eye disease. Ref-1 was highly expressed in human nAMD, murine laser-induced CNV and Vldlr-/- mouse subretinal neovascularization (SRN). Ref-1's interaction with a redox-specific small molecule inhibitor, APX2009, was shown by NMR and docking. This compound blocks crucial angiogenic features in multiple endothelial cell types. APX2009 also ameliorated murine laser-induced choroidal neovascularization (L-CNV) when delivered intravitreally. Moreover, systemic APX2009 reduced murine SRN and downregulated the expression of Ref-1 redox regulated HIF-1α target carbonic anhydrase 9 (CA9) in the Vldlr-/- mouse model. Our data validate the redox function of Ref-1 as a critical regulator of ocular angiogenesis, indicating that inhibition of Ref-1 holds therapeutic potential for treating nAMD.
Collapse
Affiliation(s)
- Anbukkarasi Muniyandi
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Gabriella D Hartman
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine, Indianapolis, IN, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN, USA
| | - Kamakshi Sishtla
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine, Indianapolis, IN, USA
- Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, ON, M5S 3M2, Canada
| | - Ratan Rai
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Cátia Gomes
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kristina Day
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Yang Song
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Andi R Masters
- Division of Clinical Pharmacology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sara K Quinney
- Division of Clinical Pharmacology, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN, USA
| | - Xiaoping Qi
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hailey Woods
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, USA
| | - Michael E Boulton
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jason S Meyer
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine, Indianapolis, IN, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jonah Z Vilseck
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Millie M Georgiadis
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN, USA
| | - Mark R Kelley
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN, USA
| | - Timothy W Corson
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA.
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine, Indianapolis, IN, USA.
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA.
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA.
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN, USA.
- Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, ON, M5S 3M2, Canada.
- Department of Ophthalmology and Vision Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
2
|
Yu J, Zhang Y, Ho M, Zhang XJ, Kam KW, Young AL, Pang CP, Tham CC, Yam JC, Chen LJ. Association of Metabolomics With Incidence of Age-Related Macular Degeneration: The UK Biobank Study. Invest Ophthalmol Vis Sci 2024; 65:43. [PMID: 39739349 DOI: 10.1167/iovs.65.14.43] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2025] Open
Abstract
Purpose The purpose of this study was to identify serum metabolites associated with age-related macular degeneration (AMD) incidence and investigate whether metabolite profiles enhance AMD risk prediction. Methods In a prospective cohort study involving 240,317 UK Biobank participants, we assessed the associations of 168 metabolites with AMD incidence using Cox hazards models. Principal component analysis (PCA) captured 90% of the variance in metabolites. These principal components (PCs) were added to the Cox models, with the first PC selected to evaluate model performance using receiver operating characteristic (ROC) curves. Results During a median follow-up of 13.69 years, 5199 (2.16%) participants developed AMD. After accounting for demographic, lifestyle, multimorbidity, socioeconomic factors, and genetic predispositions to AMD, 42 metabolites were associated with AMD incidence. Very-low-density lipoprotein (VLDL)-related particles, low-density lipoprotein (LDL)-related particles, three additional lipids particles, and albumin were associated with decreased AMD incidence, whereas glucose increased the risk of AMD incidence. Compared to those in the lowest quartile, individuals in the highest quartile of protective metabolite scores exhibited lower risk of AMD incidence (hazard ratio [HR] = 0.869, 95% confidence interval [CI] = 0.803-0.940, false discovery rate [FDR]-adjusted P = 1.44 × 10-3). However, the AMD-associated metabolites did not enhance predictive performance (both areas under the curve [AUC] = 0.776). Conclusions Our findings reveal significant associations between specific metabolites and AMD incidence, highlighting the roles of lipoprotein subclasses, cholesterol subtypes, apolipoproteins, glucose, and albumin. Although metabolomics did not improve risk prediction, certain biomarkers may serve as promising therapeutic targets.
Collapse
Affiliation(s)
- Jun Yu
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yuzhou Zhang
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Mary Ho
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
- Department of Ophthalmology and Visual Sciences, Prince of Wales Hospital, Hong Kong SAR, China
| | - Xiu Juan Zhang
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Ka Wai Kam
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
- Department of Ophthalmology and Visual Sciences, Prince of Wales Hospital, Hong Kong SAR, China
| | - Alvin L Young
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
- Department of Ophthalmology and Visual Sciences, Prince of Wales Hospital, Hong Kong SAR, China
| | - Chi Pui Pang
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Clement C Tham
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
- Department of Ophthalmology and Visual Sciences, Prince of Wales Hospital, Hong Kong SAR, China
- Hong Kong Eye Hospital, Hong Kong SAR, China
| | - Jason C Yam
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
- Hong Kong Eye Hospital, Hong Kong SAR, China
| | - Li Jia Chen
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
- Department of Ophthalmology and Visual Sciences, Prince of Wales Hospital, Hong Kong SAR, China
| |
Collapse
|
3
|
Xia CH, Liu H, Li M, Zhang H, Xing X, Gong X. Identification and Characterization of Retinitis Pigmentosa in a Novel Mouse Model Caused by PDE6B-T592I. Biomedicines 2023; 11:3173. [PMID: 38137394 PMCID: PMC10740990 DOI: 10.3390/biomedicines11123173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 11/24/2023] [Accepted: 11/27/2023] [Indexed: 12/24/2023] Open
Abstract
The cGMP-phosphodiesterase 6 beta subunit (PDE6B) is an essential component in the phototransduction pathway for light responses in photoreceptor cells. PDE6B gene mutations cause the death of rod photoreceptors, named as hereditary retinitis pigmentosa (RP) in humans and retinal degeneration (RD) in rodents. Here, we report a new RD model, identified from a phenotypic screen of N-ethyl-N-nitrosourea (ENU)-induced mutant mice, which displays retinal degeneration caused by a point mutation in the Pde6b gene that results in PDE6B-T592I mutant protein. The homozygous mutant mice show an extensive loss of rod photoreceptors at the age of 3 weeks; unexpectedly, the loss of rod photoreceptors can be partly rescued by dark rearing. Thus, this RD mutant model displays a light-dependent loss of rod photoreceptors. Both western blot and immunostaining results show very low level of mutant PDE6B-T592I protein in the retina. Structure modeling suggests that the T592I mutation probably affects the function and stability of PDE6B protein by changing intramolecular interactions. We further demonstrate that the expression of wild-type PDE6B delivered by subretinally injected adeno-associated virus (rAAV) prevents photoreceptor cell death in this RD model in vivo. The PDE6B-T592I mutant is, therefore, a valuable RD model for evaluating rAAV-mediated treatment and for investigating the molecular mechanism of light-dependent rod photoreceptor cell death that is related to impaired PDE6B function.
Collapse
Affiliation(s)
| | | | | | | | | | - Xiaohua Gong
- Herbert Wertheim School of Optometry and Vision Science Program, University of California, Berkeley, CA 94720, USA; (C.-H.X.); (H.L.); (M.L.); (H.Z.); (X.X.)
| |
Collapse
|
4
|
Bora K, Kushwah N, Maurya M, Pavlovich MC, Wang Z, Chen J. Assessment of Inner Blood-Retinal Barrier: Animal Models and Methods. Cells 2023; 12:2443. [PMID: 37887287 PMCID: PMC10605292 DOI: 10.3390/cells12202443] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 10/07/2023] [Accepted: 10/08/2023] [Indexed: 10/28/2023] Open
Abstract
Proper functioning of the neural retina relies on the unique retinal environment regulated by the blood-retinal barrier (BRB), which restricts the passage of solutes, fluids, and toxic substances. BRB impairment occurs in many retinal vascular diseases and the breakdown of BRB significantly contributes to disease pathology. Understanding the different molecular constituents and signaling pathways involved in BRB development and maintenance is therefore crucial in developing treatment modalities. This review summarizes the major molecular signaling pathways involved in inner BRB (iBRB) formation and maintenance, and representative animal models of eye diseases with retinal vascular leakage. Studies on Wnt/β-catenin signaling are highlighted, which is critical for retinal and brain vascular angiogenesis and barriergenesis. Moreover, multiple in vivo and in vitro methods for the detection and analysis of vascular leakage are described, along with their advantages and limitations. These pre-clinical animal models and methods for assessing iBRB provide valuable experimental tools in delineating the molecular mechanisms of retinal vascular diseases and evaluating therapeutic drugs.
Collapse
Affiliation(s)
| | | | | | | | | | - Jing Chen
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| |
Collapse
|
5
|
Pan XB, He YS, Lu Z, Pan HR, Wei ZY, Jin YY, Wang J, Chen JH. Epitranscriptomic investigation of myopia-associated RNA editing in the retina. Front Neurosci 2023; 17:1220114. [PMID: 37449273 PMCID: PMC10336353 DOI: 10.3389/fnins.2023.1220114] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 05/31/2023] [Indexed: 07/18/2023] Open
Abstract
Myopia is one of the most common causes of vision loss globally and is significantly affected by epigenetics. Adenosine-to-inosine (A-to-I RNA) editing is an epigenetic process involved in neurological disorders, yet its role in myopia remains undetermined. We performed a transcriptome-wide analysis of A-to-I RNA editing in the retina of form-deprivation myopia mice. Our study identified 91 A-to-I RNA editing sites in 84 genes associated with myopia. Notably, at least 27 (32.1%) of these genes with myopia-associated RNA editing showed existing evidence to be associated with myopia or related ocular phenotypes in humans or animal models, such as very low-density lipoprotein receptor (Vldlr) in retinal neovascularization and hypoxia-induced factor 1 alpha (Hif1a). Moreover, functional enrichment showed that RNA editing enriched in FDM was primarily involved in response to fungicides, a potentially druggable process for myopia prevention, and epigenetic regulation. In contrast, RNA editing enriched in controls was mostly involved in post-embryonic eye morphogenesis. Our results demonstrate altered A-to-I RNA editing associated with myopia in an experimental mouse model and warrant further study on its role in myopia development.
Collapse
Affiliation(s)
- Xu-Bin Pan
- Department of Ophthalmology, Affiliated Hospital of Jiangnan University, Wuxi, China
- Laboratory of Genomic and Precision Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
- Joint Primate Research Center for Chronic Diseases, Institute of Zoology of Guangdong Academy of Science, Jiangnan University, Wuxi, Jiangsu, China
- Jiangnan University Brain Institute, Wuxi, Jiangsu, China
| | - Yu-Shan He
- Laboratory of Genomic and Precision Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
- Joint Primate Research Center for Chronic Diseases, Institute of Zoology of Guangdong Academy of Science, Jiangnan University, Wuxi, Jiangsu, China
- Jiangnan University Brain Institute, Wuxi, Jiangsu, China
| | - Zijing Lu
- Department of Ophthalmology, Affiliated Hospital of Jiangnan University, Wuxi, China
- Laboratory of Genomic and Precision Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
- Joint Primate Research Center for Chronic Diseases, Institute of Zoology of Guangdong Academy of Science, Jiangnan University, Wuxi, Jiangsu, China
- Jiangnan University Brain Institute, Wuxi, Jiangsu, China
| | - Hao-Ran Pan
- Department of Ophthalmology, Affiliated Hospital of Jiangnan University, Wuxi, China
- Laboratory of Genomic and Precision Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
- Joint Primate Research Center for Chronic Diseases, Institute of Zoology of Guangdong Academy of Science, Jiangnan University, Wuxi, Jiangsu, China
- Jiangnan University Brain Institute, Wuxi, Jiangsu, China
| | - Zhi-Yuan Wei
- Laboratory of Genomic and Precision Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
- Joint Primate Research Center for Chronic Diseases, Institute of Zoology of Guangdong Academy of Science, Jiangnan University, Wuxi, Jiangsu, China
- Jiangnan University Brain Institute, Wuxi, Jiangsu, China
| | - Yun-Yun Jin
- Laboratory of Genomic and Precision Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
- Joint Primate Research Center for Chronic Diseases, Institute of Zoology of Guangdong Academy of Science, Jiangnan University, Wuxi, Jiangsu, China
- Jiangnan University Brain Institute, Wuxi, Jiangsu, China
| | - Jihong Wang
- Department of Ophthalmology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Jian-Huan Chen
- Laboratory of Genomic and Precision Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
- Joint Primate Research Center for Chronic Diseases, Institute of Zoology of Guangdong Academy of Science, Jiangnan University, Wuxi, Jiangsu, China
- Jiangnan University Brain Institute, Wuxi, Jiangsu, China
| |
Collapse
|
6
|
Forward genetic analysis using OCT screening identifies Sfxn3 mutations leading to progressive outer retinal degeneration in mice. Proc Natl Acad Sci U S A 2020; 117:12931-12942. [PMID: 32457148 DOI: 10.1073/pnas.1921224117] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Retinal disease and loss of vision can result from any disruption of the complex pathways controlling retinal development and homeostasis. Forward genetics provides an excellent tool to find, in an unbiased manner, genes that are essential to these processes. Using N-ethyl-N-nitrosourea mutagenesis in mice in combination with a screening protocol using optical coherence tomography (OCT) and automated meiotic mapping, we identified 11 mutations presumably causative of retinal phenotypes in genes previously known to be essential for retinal integrity. In addition, we found multiple statistically significant gene-phenotype associations that have not been reported previously and decided to target one of these genes, Sfxn3 (encoding sideroflexin-3), using CRISPR/Cas9 technology. We demonstrate, using OCT, light microscopy, and electroretinography, that two Sfxn3 -/- mouse lines developed progressive and severe outer retinal degeneration. Electron microscopy showed thinning of the retinal pigment epithelium and disruption of the external limiting membrane. Using single-cell RNA sequencing of retinal cells isolated from C57BL/6J mice, we demonstrate that Sfxn3 is expressed in several bipolar cell subtypes, retinal ganglion cells, and some amacrine cell subtypes but not significantly in Müller cells or photoreceptors. In situ hybridization confirmed these findings. Furthermore, pathway analysis suggests that Sfxn3 may be associated with synaptic homeostasis. Importantly, electron microscopy analysis showed disruption of synapses and synaptic ribbons in the outer plexiform layer of Sfxn3 -/- mice. Our work describes a previously unknown requirement for Sfxn3 in retinal function.
Collapse
|
7
|
Collin GB, Gogna N, Chang B, Damkham N, Pinkney J, Hyde LF, Stone L, Naggert JK, Nishina PM, Krebs MP. Mouse Models of Inherited Retinal Degeneration with Photoreceptor Cell Loss. Cells 2020; 9:cells9040931. [PMID: 32290105 PMCID: PMC7227028 DOI: 10.3390/cells9040931] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 04/05/2020] [Accepted: 04/07/2020] [Indexed: 12/12/2022] Open
Abstract
Inherited retinal degeneration (RD) leads to the impairment or loss of vision in millions of individuals worldwide, most frequently due to the loss of photoreceptor (PR) cells. Animal models, particularly the laboratory mouse, have been used to understand the pathogenic mechanisms that underlie PR cell loss and to explore therapies that may prevent, delay, or reverse RD. Here, we reviewed entries in the Mouse Genome Informatics and PubMed databases to compile a comprehensive list of monogenic mouse models in which PR cell loss is demonstrated. The progression of PR cell loss with postnatal age was documented in mutant alleles of genes grouped by biological function. As anticipated, a wide range in the onset and rate of cell loss was observed among the reported models. The analysis underscored relationships between RD genes and ciliary function, transcription-coupled DNA damage repair, and cellular chloride homeostasis. Comparing the mouse gene list to human RD genes identified in the RetNet database revealed that mouse models are available for 40% of the known human diseases, suggesting opportunities for future research. This work may provide insight into the molecular players and pathways through which PR degenerative disease occurs and may be useful for planning translational studies.
Collapse
Affiliation(s)
- Gayle B. Collin
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; (G.B.C.); (N.G.); (B.C.); (N.D.); (J.P.); (L.F.H.); (L.S.); (J.K.N.)
| | - Navdeep Gogna
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; (G.B.C.); (N.G.); (B.C.); (N.D.); (J.P.); (L.F.H.); (L.S.); (J.K.N.)
| | - Bo Chang
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; (G.B.C.); (N.G.); (B.C.); (N.D.); (J.P.); (L.F.H.); (L.S.); (J.K.N.)
| | - Nattaya Damkham
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; (G.B.C.); (N.G.); (B.C.); (N.D.); (J.P.); (L.F.H.); (L.S.); (J.K.N.)
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Jai Pinkney
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; (G.B.C.); (N.G.); (B.C.); (N.D.); (J.P.); (L.F.H.); (L.S.); (J.K.N.)
| | - Lillian F. Hyde
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; (G.B.C.); (N.G.); (B.C.); (N.D.); (J.P.); (L.F.H.); (L.S.); (J.K.N.)
| | - Lisa Stone
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; (G.B.C.); (N.G.); (B.C.); (N.D.); (J.P.); (L.F.H.); (L.S.); (J.K.N.)
| | - Jürgen K. Naggert
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; (G.B.C.); (N.G.); (B.C.); (N.D.); (J.P.); (L.F.H.); (L.S.); (J.K.N.)
| | - Patsy M. Nishina
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; (G.B.C.); (N.G.); (B.C.); (N.D.); (J.P.); (L.F.H.); (L.S.); (J.K.N.)
- Correspondence: (P.M.N.); (M.P.K.); Tel.: +1-207-2886-383 (P.M.N.); +1-207-2886-000 (M.P.K.)
| | - Mark P. Krebs
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; (G.B.C.); (N.G.); (B.C.); (N.D.); (J.P.); (L.F.H.); (L.S.); (J.K.N.)
- Correspondence: (P.M.N.); (M.P.K.); Tel.: +1-207-2886-383 (P.M.N.); +1-207-2886-000 (M.P.K.)
| |
Collapse
|
8
|
Qiu F, Matlock G, Chen Q, Zhou K, Du Y, Wang X, Ma JX. Therapeutic Effects of PPARα Agonist on Ocular Neovascularization in Models Recapitulating Neovascular Age-Related Macular Degeneration. Invest Ophthalmol Vis Sci 2017; 58:5065-5075. [PMID: 28980001 PMCID: PMC5633006 DOI: 10.1167/iovs.17-22091] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Purpose This study was designed to evaluate effects of fenofibric acid (Feno-FA), a peroxisome proliferator–activated receptor-alpha (PPARα) agonist, on ocular neovascularization (NV) in models recapitulating neovascular age-related macular degeneration (AMD), and to explore whether the effects are PPARα dependent. Methods Laser-induced choroidal NV (CNV) in rats and very low-density lipoprotein receptor knockout (Vldlr−/−) mice received daily intraperitoneal injections of Feno-FA or vehicle. Vascular leakage was examined by fundus fluorescein angiography and permeability assay using Evans blue as tracer. In CNV rats, severity of CNV was evaluated by CNV areas and CNV volume. In Vldlr−/− mice, subretinal NV (SRNV) and intraretinal NV (IRNV) were quantified in choroid flat mount and retina flat mount, respectively. Inflammatory factors were measured using Western blotting and retinal leukostasis assay. Further, Pparα−/− mice and age-matched wild-type (WT) mice were used for laser-induced CNV and treated with Feno-FA to explore the underlying mechanism. Results Feno-FA significantly reduced vascular leakage in CNV rats and Vldlr−/− mice, reduced CNV volume in laser-induced CNV rats, and suppressed SRNV and IRNV in Vldlr−/− mice. In addition, Feno-FA downregulated the expression of inflammatory factors, including VEGF, TNF-α, and intercellular cell adhesion molecule-1 (ICAM-1), in the eyecups of CNV rats and decreased adherent retinal leukocytes in Vldlr−/− mice. Furthermore, Pparα−/− mice developed more severe CNV compared with WT mice, and PPARα knockout abolished the beneficial effects of Feno-FA on CNV. Conclusions Feno-FA has therapeutic effects on ocular NV in models recapitulating neovascular AMD through a PPARα-dependent mechanism.
Collapse
Affiliation(s)
- Fangfang Qiu
- Department of Physiology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Greg Matlock
- Department of Physiology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Qian Chen
- Department of Physiology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Kelu Zhou
- Department of Physiology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Yanhong Du
- Department of Physiology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Xiang Wang
- Department of Cell Biology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Jian-Xing Ma
- Department of Physiology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| |
Collapse
|
9
|
Cheung CMG, Gan A, Fan Q, Chee ML, Apte RS, Khor CC, Yeo I, Mathur R, Cheng CY, Wong TY, Tai ES. Plasma lipoprotein subfraction concentrations are associated with lipid metabolism and age-related macular degeneration. J Lipid Res 2017; 58:1785-1796. [PMID: 28698208 PMCID: PMC5580892 DOI: 10.1194/jlr.m073684] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2016] [Revised: 07/06/2017] [Indexed: 01/09/2023] Open
Abstract
Disturbance in lipid metabolism has been suggested as a major pathogenic factor for age-related macular degeneration (AMD). Conventional lipid measures have been inconsistently associated with AMD. Other factors that can alter lipid metabolism include lipoprotein phenotype and genetic mutations. We performed a case-control study to examine the association between lipoprotein profile and neovascular AMD (nAMD) and whether the cholesterylester transfer protein (CETP) D442G mutation modulates these associations. Patients with nAMD had significantly higher concentrations of HDL and IDL compared with controls. The increase in HDL particles in nAMD patients was driven by an excess of medium-sized particles. Concurrently, patients with nAMD also had lower Apo A-1, lower VLDL and chylomicron lipoprotein. Many of these associations showed a dose-dependent association between controls, early AMD cases, and nAMD cases. Adjustment for the presence of the D442G mutation at the CETP locus did not significantly alter the increased AMD risk associated with HDL particle concentration. AMD is associated with variation in many lipoprotein subclasses, including increased HDL and IDL particles and decreased Apo A-1, VLDL, and chylomicron particles. These data suggest widespread systemic disturbance in lipid metabolism in the pathogenesis of AMD, including possible alterations in lipoprotein carrier capacity.
Collapse
Affiliation(s)
- Chui Ming Gemmy Cheung
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore; Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Ophthalmology and Visual Sciences Program, Duke-NUS Medical School, National University of Singapore, Singapore.
| | - Alfred Gan
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
| | - Qiao Fan
- Centre for Quantitative Medicine, Duke-NUS Medical School, National University of Singapore, Singapore
| | - Miao Ling Chee
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
| | - Rajendra S Apte
- Ophthalmology and Visual Sciences, Developmental Biology and Medicine, Washington University School of Medicine, St. Louis, MO
| | | | - Ian Yeo
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore; Ophthalmology and Visual Sciences Program, Duke-NUS Medical School, National University of Singapore, Singapore
| | - Ranjana Mathur
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore; Ophthalmology and Visual Sciences Program, Duke-NUS Medical School, National University of Singapore, Singapore
| | - Ching-Yu Cheng
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore; Ophthalmology and Visual Sciences Program, Duke-NUS Medical School, National University of Singapore, Singapore
| | - Tien Yin Wong
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore; Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Ophthalmology and Visual Sciences Program, Duke-NUS Medical School, National University of Singapore, Singapore
| | - E Shyong Tai
- Department of Medicine, Cardiovascular and Metabolic Disorders Programme, National University of Singapore, Singapore
| |
Collapse
|
10
|
Housset M, Sennlaub F. Thrombospondin-1 and Pathogenesis of Age-Related Macular Degeneration. J Ocul Pharmacol Ther 2015; 31:406-12. [DOI: 10.1089/jop.2015.0023] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
- Michael Housset
- Sorbonne Universités, UPMC Univ Paris 06, UMR_S 968, Institut de la Vision, Paris, France
- CNRS, UMR_7210, Paris, France
- INSERM, U968, Paris, France
| | - Florian Sennlaub
- Sorbonne Universités, UPMC Univ Paris 06, UMR_S 968, Institut de la Vision, Paris, France
- CNRS, UMR_7210, Paris, France
- INSERM, U968, Paris, France
- Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, DHU ViewMaintain, INSERM-DHOS CIC 1423, Paris, France
| |
Collapse
|
11
|
Johnson V, Xiang M, Chen Z, Junge HJ. Neurite Mistargeting and Inverse Order of Intraretinal Vascular Plexus Formation Precede Subretinal Vascularization in Vldlr Mutant Mice. PLoS One 2015; 10:e0132013. [PMID: 26177550 PMCID: PMC4503745 DOI: 10.1371/journal.pone.0132013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 06/09/2015] [Indexed: 02/07/2023] Open
Abstract
In the retina blood vessels are required to support a high metabolic rate, however, uncontrolled vascular growth can lead to impaired vision and blindness. Subretinal vascularization (SRV), one type of pathological vessel growth, occurs in retinal angiomatous proliferation and proliferative macular telangiectasia. In these diseases SRV originates from blood vessels within the retina. We use mice with a targeted disruption in the Vldl-receptor (Vldlr) gene as a model to study SRV with retinal origin. We find that Vldlr mRNA is strongly expressed in the neuroretina, and we observe both vascular and neuronal phenotypes in Vldlr-/- mice. Unexpectedly, horizontal cell (HC) neurites are mistargeted prior to SRV in this model, and the majority of vascular lesions are associated with mistargeted neurites. In Foxn4-/- mice, which lack HCs and display reduced amacrine cell (AC) numbers, we find severe defects in intraretinal capillary development. However, SRV is not suppressed in Foxn4-/-;Vldlr-/- mice, which reveals that mistargeted HC neurites are not required for vascular lesion formation. In the absence of VLDLR, the intraretinal capillary plexuses form in an inverse order compared to normal development, and subsequent to this early defect, vascular proliferation is increased. We conclude that SRV in the Vldlr-/- model is associated with mistargeted neurites and that SRV is preceded by altered retinal vascular development.
Collapse
Affiliation(s)
- Verity Johnson
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, Colorado, 80309, United States of America
| | - Mengqing Xiang
- Center for Advanced Biotechnology and Medicine and Department of Pediatrics, Rutgers University-Robert Wood Johnson Medical School, Piscataway, New Jersey, 08901, United States of America
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, 54 South Xianlie Road, Guangzhou, 510060, China
| | - Zhe Chen
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, Colorado, 80309, United States of America
| | - Harald J. Junge
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, Colorado, 80309, United States of America
| |
Collapse
|
12
|
Wu J, Strawn TL, Luo M, Wang L, Li R, Ren M, Xia J, Zhang Z, Ma W, Luo T, Lawrence DA, Fay WP. Plasminogen activator inhibitor-1 inhibits angiogenic signaling by uncoupling vascular endothelial growth factor receptor-2-αVβ3 integrin cross talk. Arterioscler Thromb Vasc Biol 2015; 35:111-20. [PMID: 25378411 PMCID: PMC4270947 DOI: 10.1161/atvbaha.114.304554] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Accepted: 10/25/2014] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Plasminogen activator inhibitor-1 (PAI-1) regulates angiogenesis via effects on extracellular matrix proteolysis and cell adhesion. However, no previous study has implicated PAI-1 in controlling vascular endothelial growth factor (VEGF) signaling. We tested the hypothesis that PAI-1 downregulates VEGF receptor-2 (VEGFR-2) activation by inhibiting a vitronectin-dependent cooperative binding interaction between VEGFR-2 and αVβ3. APPROACH AND RESULTS We studied effects of PAI-1 on VEGF signaling in human umbilical vein endothelial cells. PAI-1 inhibited VEGF-induced phosphorylation of VEGFR-2 in human umbilical vein endothelial cells grown on vitronectin, but not on fibronectin or collagen. PAI-1 inhibited the binding of VEGFR-2 to β3 integrin, VEGFR-2 endocytosis, and intracellular signaling pathways downstream of VEGFR-2. The anti-VEGF effect of PAI-1 was mediated by 2 distinct pathways, one requiring binding to vitronectin and another requiring binding to very low-density lipoprotein receptor. PAI-1 inhibited VEGF-induced angiogenesis in vitro and in vivo, and pharmacological inhibition of PAI-1 promoted collateral arteriole development and recovery of hindlimb perfusion after femoral artery interruption. CONCLUSIONS PAI-1 inhibits activation of VEGFR-2 by VEGF by disrupting a vitronectin-dependent proangiogenic binding interaction involving αVβ3 and VEGFR-2. These results broaden our understanding of the roles of PAI-1, vitronectin, and endocytic receptors in regulating VEGFR-2 activation and suggest novel therapeutic strategies for regulating VEGF signaling.
Collapse
Affiliation(s)
- Jianbo Wu
- From the Departments of Internal Medicine and Medical Pharmacology and Physiology (J.W., T.L.S., M.L., W.P.F.), the Research Service, Harry S. Truman Memorial Veterans Hospital (W.P.F.), University of Missouri School of Medicine, Columbia; the Drug Discovery Research Center, Luzhou Medical College, Luzhou, Sichuan, China (J.W., M.L., L.W., R.L., M.R., J.X., Z.Z., W.M., T.L.); and the Department of Internal Medicine, University of Michigan Medical School, Ann Arbor (D.A.L.).
| | - Tammy L Strawn
- From the Departments of Internal Medicine and Medical Pharmacology and Physiology (J.W., T.L.S., M.L., W.P.F.), the Research Service, Harry S. Truman Memorial Veterans Hospital (W.P.F.), University of Missouri School of Medicine, Columbia; the Drug Discovery Research Center, Luzhou Medical College, Luzhou, Sichuan, China (J.W., M.L., L.W., R.L., M.R., J.X., Z.Z., W.M., T.L.); and the Department of Internal Medicine, University of Michigan Medical School, Ann Arbor (D.A.L.)
| | - Mao Luo
- From the Departments of Internal Medicine and Medical Pharmacology and Physiology (J.W., T.L.S., M.L., W.P.F.), the Research Service, Harry S. Truman Memorial Veterans Hospital (W.P.F.), University of Missouri School of Medicine, Columbia; the Drug Discovery Research Center, Luzhou Medical College, Luzhou, Sichuan, China (J.W., M.L., L.W., R.L., M.R., J.X., Z.Z., W.M., T.L.); and the Department of Internal Medicine, University of Michigan Medical School, Ann Arbor (D.A.L.)
| | - Liqun Wang
- From the Departments of Internal Medicine and Medical Pharmacology and Physiology (J.W., T.L.S., M.L., W.P.F.), the Research Service, Harry S. Truman Memorial Veterans Hospital (W.P.F.), University of Missouri School of Medicine, Columbia; the Drug Discovery Research Center, Luzhou Medical College, Luzhou, Sichuan, China (J.W., M.L., L.W., R.L., M.R., J.X., Z.Z., W.M., T.L.); and the Department of Internal Medicine, University of Michigan Medical School, Ann Arbor (D.A.L.)
| | - Rong Li
- From the Departments of Internal Medicine and Medical Pharmacology and Physiology (J.W., T.L.S., M.L., W.P.F.), the Research Service, Harry S. Truman Memorial Veterans Hospital (W.P.F.), University of Missouri School of Medicine, Columbia; the Drug Discovery Research Center, Luzhou Medical College, Luzhou, Sichuan, China (J.W., M.L., L.W., R.L., M.R., J.X., Z.Z., W.M., T.L.); and the Department of Internal Medicine, University of Michigan Medical School, Ann Arbor (D.A.L.)
| | - Meiping Ren
- From the Departments of Internal Medicine and Medical Pharmacology and Physiology (J.W., T.L.S., M.L., W.P.F.), the Research Service, Harry S. Truman Memorial Veterans Hospital (W.P.F.), University of Missouri School of Medicine, Columbia; the Drug Discovery Research Center, Luzhou Medical College, Luzhou, Sichuan, China (J.W., M.L., L.W., R.L., M.R., J.X., Z.Z., W.M., T.L.); and the Department of Internal Medicine, University of Michigan Medical School, Ann Arbor (D.A.L.)
| | - Jiyi Xia
- From the Departments of Internal Medicine and Medical Pharmacology and Physiology (J.W., T.L.S., M.L., W.P.F.), the Research Service, Harry S. Truman Memorial Veterans Hospital (W.P.F.), University of Missouri School of Medicine, Columbia; the Drug Discovery Research Center, Luzhou Medical College, Luzhou, Sichuan, China (J.W., M.L., L.W., R.L., M.R., J.X., Z.Z., W.M., T.L.); and the Department of Internal Medicine, University of Michigan Medical School, Ann Arbor (D.A.L.)
| | - Zhuo Zhang
- From the Departments of Internal Medicine and Medical Pharmacology and Physiology (J.W., T.L.S., M.L., W.P.F.), the Research Service, Harry S. Truman Memorial Veterans Hospital (W.P.F.), University of Missouri School of Medicine, Columbia; the Drug Discovery Research Center, Luzhou Medical College, Luzhou, Sichuan, China (J.W., M.L., L.W., R.L., M.R., J.X., Z.Z., W.M., T.L.); and the Department of Internal Medicine, University of Michigan Medical School, Ann Arbor (D.A.L.)
| | - Weizhong Ma
- From the Departments of Internal Medicine and Medical Pharmacology and Physiology (J.W., T.L.S., M.L., W.P.F.), the Research Service, Harry S. Truman Memorial Veterans Hospital (W.P.F.), University of Missouri School of Medicine, Columbia; the Drug Discovery Research Center, Luzhou Medical College, Luzhou, Sichuan, China (J.W., M.L., L.W., R.L., M.R., J.X., Z.Z., W.M., T.L.); and the Department of Internal Medicine, University of Michigan Medical School, Ann Arbor (D.A.L.)
| | - Tingting Luo
- From the Departments of Internal Medicine and Medical Pharmacology and Physiology (J.W., T.L.S., M.L., W.P.F.), the Research Service, Harry S. Truman Memorial Veterans Hospital (W.P.F.), University of Missouri School of Medicine, Columbia; the Drug Discovery Research Center, Luzhou Medical College, Luzhou, Sichuan, China (J.W., M.L., L.W., R.L., M.R., J.X., Z.Z., W.M., T.L.); and the Department of Internal Medicine, University of Michigan Medical School, Ann Arbor (D.A.L.)
| | - Daniel A Lawrence
- From the Departments of Internal Medicine and Medical Pharmacology and Physiology (J.W., T.L.S., M.L., W.P.F.), the Research Service, Harry S. Truman Memorial Veterans Hospital (W.P.F.), University of Missouri School of Medicine, Columbia; the Drug Discovery Research Center, Luzhou Medical College, Luzhou, Sichuan, China (J.W., M.L., L.W., R.L., M.R., J.X., Z.Z., W.M., T.L.); and the Department of Internal Medicine, University of Michigan Medical School, Ann Arbor (D.A.L.)
| | - William P Fay
- From the Departments of Internal Medicine and Medical Pharmacology and Physiology (J.W., T.L.S., M.L., W.P.F.), the Research Service, Harry S. Truman Memorial Veterans Hospital (W.P.F.), University of Missouri School of Medicine, Columbia; the Drug Discovery Research Center, Luzhou Medical College, Luzhou, Sichuan, China (J.W., M.L., L.W., R.L., M.R., J.X., Z.Z., W.M., T.L.); and the Department of Internal Medicine, University of Michigan Medical School, Ann Arbor (D.A.L.)
| |
Collapse
|
13
|
Yang D, Gao L, Wang T, Qiao Z, Liang Y, Zhang P. Hypoxia triggers endothelial endoplasmic reticulum stress and apoptosis via induction of VLDL receptor. FEBS Lett 2014; 588:4448-56. [DOI: 10.1016/j.febslet.2014.09.046] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Revised: 09/10/2014] [Accepted: 09/16/2014] [Indexed: 01/10/2023]
|
14
|
Deletion of LRP5 in VLDLR knockout mice inhibits retinal neovascularization. PLoS One 2013; 8:e75186. [PMID: 24058663 PMCID: PMC3772893 DOI: 10.1371/journal.pone.0075186] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Accepted: 08/12/2013] [Indexed: 11/19/2022] Open
Abstract
The development and maintenance of retinal vasculature require a precise balance between pro-angiogenic and anti-angiogenic factors. However, mechanisms underlying normal homeostasis of retinal vasculature and pathological changes of disrupted retinal vessel development are not fully understood. Recent studies of the low-density lipoprotein receptor-related protein 5 (LRP5) and the very low-density lipoprotein receptor (VLDLR) mutant mice indicate that LRP5 mediates a pro-angiogenic signal while VLDLR mediates an anti-angiogenic signal in retinal vasculature. Mice with a loss of LRP5 display underdeveloped intraretinal vasculature associated with endothelial cell (EC) clustering and failed EC migration into deep retinal layers. In contrast, VLDLR knockout mice show overgrown intraretinal vasculature and subretinal neovascularization. To understand the mechanisms for the opposite retinal vascular abnormalities between LRP5 and VLDLR mutant mice and to test how a loss of LRP5 perturbs subretinal neovascularization caused by a loss of VLDLR, we have generated and characterized the retinal vasculature in LRP5/VLDLR double knockout (DKO) mice. Our data show that DKO mice develop substantial EC clustering without subretinal neovascularization. The absence of subretinal neovascularization in DKO mice is associated with inhibited migration of ECs into the photoreceptor cell layer. In addition, the transcription level of Slc38a5, which encodes a Müller cell specific glutamine transporter, is significantly reduced in DKO mice, similar to previously reported changes in LRP5 single knockout mice. Thus, LRP5 signaling is a prerequisite for neovascularization in VLDLR knockout mice. LRP5 may be an effective target for inhibiting intraretinal neovascularization.
Collapse
|
15
|
Kyosseva SV, Chen L, Seal S, McGinnis JF. Nanoceria inhibit expression of genes associated with inflammation and angiogenesis in the retina of Vldlr null mice. Exp Eye Res 2013; 116:63-74. [PMID: 23978600 DOI: 10.1016/j.exer.2013.08.003] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Revised: 07/02/2013] [Accepted: 08/06/2013] [Indexed: 12/26/2022]
Abstract
Oxidative stress and inflammation are important pathological mechanisms in many neurodegenerative diseases, including age-related macular degeneration (AMD). The very low-density lipoprotein receptor knockout mouse (Vldlr-/-) has been identified as a model for AMD and in particular for retinal angiomatous proliferation (RAP). In this study we examined the effect of cerium oxide nanoparticles (nanoceria) that have been shown to have catalytic antioxidant activity, on expression of 88 major cytokines in the retinas of Vldlr-/- mice using a PCR array. A single intravitreal injection of nanoceria at P28 caused inhibition of pro-inflammatory cytokines and pro-angiogenic growth factors including Tslp, Lif, Il3, Il7, Vegfa, Fgf1, Fgf2, Fgf7, Egf, Efna3, Lep, and up-regulation of several cytokines and anti-angiogenic genes in the Vldlr-/- retina within one week. We used the Ingenuity Pathway Analysis software to search for biological functions, pathways, and interrelationships between gene networks. Many of the genes whose activities were affected are involved in cell signaling, cellular development, growth and proliferation, and tissue development. Western blot analysis revealed that nanoceria inhibit the activation of ERK 1/2, JNK, p38 MAP kinase, and Akt. These data suggest that nanoceria may represent a novel therapeutic strategy to treat AMD, RAP, and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Svetlana V Kyosseva
- Department of Ophthalmology/Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.
| | | | | | | |
Collapse
|
16
|
Weinl C, Riehle H, Park D, Stritt C, Beck S, Huber G, Wolburg H, Olson EN, Seeliger MW, Adams RH, Nordheim A. Endothelial SRF/MRTF ablation causes vascular disease phenotypes in murine retinae. J Clin Invest 2013; 123:2193-206. [PMID: 23563308 DOI: 10.1172/jci64201] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2012] [Accepted: 02/07/2013] [Indexed: 11/17/2022] Open
Abstract
Retinal vessel homeostasis ensures normal ocular functions. Consequently, retinal hypovascularization and neovascularization, causing a lack and an excess of vessels, respectively, are hallmarks of human retinal pathology. We provide evidence that EC-specific genetic ablation of either the transcription factor SRF or its cofactors MRTF-A and MRTF-B, but not the SRF cofactors ELK1 or ELK4, cause retinal hypovascularization in the postnatal mouse eye. Inducible, EC-specific deficiency of SRF or MRTF-A/MRTF-B during postnatal angiogenesis impaired endothelial tip cell filopodia protrusion, resulting in incomplete formation of the retinal primary vascular plexus, absence of the deep plexi, and persistence of hyaloid vessels. All of these features are typical of human hypovascularization-related vitreoretinopathies, such as familial exudative vitreoretinopathies including Norrie disease. In contrast, conditional EC deletion of Srf in adult murine vessels elicited intraretinal neovascularization that was reminiscent of the age-related human pathologies retinal angiomatous proliferation and macular telangiectasia. These results indicate that angiogenic homeostasis is ensured by differential stage-specific functions of SRF target gene products in the developing versus the mature retinal vasculature and suggest that the actin-directed MRTF-SRF signaling axis could serve as a therapeutic target in the treatment of human vascular retinal diseases.
Collapse
Affiliation(s)
- Christine Weinl
- Department for Molecular Biology, Interfaculty Institute of Cell Biology, University of Tübingen, Tübingen, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|