1
|
Shi X, Xue Z, Ye K, Yuan J, Zhang Y, Qu J, Su J. Roles of non-coding RNAs in eye development and diseases. WILEY INTERDISCIPLINARY REVIEWS. RNA 2023; 14:e1785. [PMID: 36849659 DOI: 10.1002/wrna.1785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 12/17/2022] [Accepted: 02/06/2023] [Indexed: 03/01/2023]
Abstract
The prevalence of ocular disorders is dramatically increasing worldwide, especially those that cause visual impairment and permanent loss of vision, including cataract, glaucoma, age-related macular degeneration, and diabetic retinopathy. Extensive evidence has shown that ncRNAs are key regulators in various biogenesis and biological functions, controlling gene expression related to histogenesis and cell differentiation in ocular tissues. Aberrant expression and function of ncRNA can lead to dysfunction of visual system and mediate progression of eye disorders. Here, we mainly offer an overview of the role of precise modulation of ncRNAs in eye development and function in patients with eye diseases. We also highlight the challenges and future perspectives in conducting ncRNA studies, focusing specifically on the role of ncRNAs that may hold expanded promise for their diagnostic and therapeutic applications in various eye diseases. This article is categorized under: Regulatory RNAs/RNAi/Riboswitches > Regulatory RNAs RNA in Disease and Development > RNA in Disease RNA in Disease and Development > RNA in Development.
Collapse
Affiliation(s)
- Xinrui Shi
- School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhengbo Xue
- School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Kaicheng Ye
- School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jian Yuan
- School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Zhejiang, China
| | - Yan Zhang
- School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jia Qu
- School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Zhejiang, China
| | - Jianzhong Su
- School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Zhejiang, China
- Institute of PSI Genomics, Zhejiang, China
| |
Collapse
|
2
|
Davari M, Soheili ZS, Latifi-Navid H, Samiee S. Potential involvement of miR-183/96/182 cluster-gene target interactions in transdifferentiation of human retinal pigment epithelial cells into retinal neurons. Biochem Biophys Res Commun 2023; 663:87-95. [PMID: 37119770 DOI: 10.1016/j.bbrc.2023.04.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 04/12/2023] [Accepted: 04/16/2023] [Indexed: 05/01/2023]
Abstract
miR-183/96/182 cluster plays a critical role in the developing retina by regulating many target genes involved in signaling pathways. This study aimed to survey the miR-183/96/182 cluster-target interactions that, potentially contribute to human retinal pigmented epithelial (hRPE) cell differentiation into photoreceptors. Target genes of the miR-183/96/182 cluster were obtained from miRNA-target databases and applied to construct miRNA-target networks. Gene ontology and KEGG pathway analysis was performed. miR-183/96/182 cluster sequence was cloned into an eGFP-intron splicing cassette in an AAV2 vector and overexpressed in hRPE cells. The expression level of target genes including HES1, PAX6, SOX2, CCNJ, and RORΒ was evaluated using qPCR. Our results showed that miR-183, miR-96, and miR-182 share 136 target genes that are involved in cell proliferation pathways such as PI3K/AKT and MAPK pathway. qPCR data indicated a 22-, 7-, and 4-fold overexpression of miR-183, miR-96, and miR-182, respectively, in infected hRPE cells. Consequently, the downregulation of several key targets such as PAX6, CCND2, CDK5R1, and CCNJ and upregulation of a few retina-specific neural markers such as Rhodopsin, red opsin, and CRX was detected. Our findings suggest that the miR-183/96/182 cluster may induce hRPE transdifferentiation by targeting key genes that involve in the cell cycle and proliferation pathways.
Collapse
Affiliation(s)
- Maliheh Davari
- Molecular Medicine Department, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Zahra-Soheila Soheili
- Molecular Medicine Department, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran.
| | - Hamid Latifi-Navid
- Molecular Medicine Department, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Shahram Samiee
- Blood Transfusion Research Center High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| |
Collapse
|
3
|
Navarro-Calvo J, Esquiva G, Gómez-Vicente V, Valor LM. MicroRNAs in the Mouse Developing Retina. Int J Mol Sci 2023; 24:ijms24032992. [PMID: 36769311 PMCID: PMC9918188 DOI: 10.3390/ijms24032992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/23/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
The retina is among the highest organized tissues of the central nervous system. To achieve such organization, a finely tuned regulation of developmental processes is required to form the retinal layers that contain the specialized neurons and supporting glial cells to allow precise phototransduction. MicroRNAs are a class of small RNAs with undoubtful roles in fundamental biological processes, including neurodevelopment of the brain and the retina. This review provides a short overview of the most important findings regarding microRNAs in the regulation of retinal development, from the developmental-dependent rearrangement of the microRNA expression program to the key roles of particular microRNAs in the differentiation and maintenance of retinal cell subtypes.
Collapse
Affiliation(s)
- Jorge Navarro-Calvo
- Unidad de Investigación, Hospital General Universitario Dr. Balmis, ISABIAL, 03010 Alicante, Spain
| | - Gema Esquiva
- Department of Optics, Pharmacology and Anatomy, University of Alicante, 03690 Alicante, Spain
| | - Violeta Gómez-Vicente
- Department of Optics, Pharmacology and Anatomy, University of Alicante, 03690 Alicante, Spain
| | - Luis M. Valor
- Unidad de Investigación, Hospital General Universitario Dr. Balmis, ISABIAL, 03010 Alicante, Spain
- Correspondence: ; Tel.: +34-965-913-988
| |
Collapse
|
4
|
Lucci C, De Groef L. On the other end of the line: Extracellular vesicle-mediated communication in glaucoma. Front Neuroanat 2023; 17:1148956. [PMID: 37113676 PMCID: PMC10126352 DOI: 10.3389/fnana.2023.1148956] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 03/27/2023] [Indexed: 04/29/2023] Open
Abstract
In the last decade, extracellular vesicles (EVs) have emerged as a promising field of research due to their ability to participate in cell-to-cell communication via the transfer of their very diverse and complex cargo. The latter reflects the nature and physiological state of the cell of origin and, as such, EVs may not only play a pivotal role in the cellular events that culminate into disease, but also hold great potential as drug delivery vehicles and biomarkers. Yet, their role in glaucoma, the leading cause of irreversible blindness worldwide, has not been fully studied. Here, we provide an overview of the different EV subtypes along with their biogenesis and content. We elaborate on how EVs released by different cell types can exert a specific function in the context of glaucoma. Finally, we discuss how these EVs provide opportunities to be used as biomarkers for diagnosis and monitoring of disease.
Collapse
|
5
|
Kadir RRA, Alwjwaj M, Bayraktutan U. MicroRNA: An Emerging Predictive, Diagnostic, Prognostic and Therapeutic Strategy in Ischaemic Stroke. Cell Mol Neurobiol 2022; 42:1301-1319. [PMID: 33368054 PMCID: PMC9142420 DOI: 10.1007/s10571-020-01028-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 12/09/2020] [Indexed: 02/07/2023]
Abstract
Stroke continues to be the third-leading cause of death and disability worldwide. The limited availability of diagnostic tools approved therapeutics and biomarkers that help monitor disease progression or predict future events remain as the major challenges in the field of stroke medicine. Hence, attempts to discover safe and efficacious therapeutics and reliable biomarkers are of paramount importance. MicroRNAs (miRNAs) are a class of non-coding RNAs that play important roles in regulating gene expression. Since miRNAs also play important roles in key mechanisms associated with the pathogenesis of stroke, including energy failure, inflammation and cell death, it is possible that miRNAs may serve as reliable blood-based markers for risk prediction, diagnosis and prognosis of ischaemic stroke. Discovery of better neurological outcome and smaller cerebral infarcts in animal models of ischaemic stroke treated with miRNA agomirs or antagomirs indicate that miRNAs may also play a cerebrovascular protective role after an ischaemic stroke. Nonetheless, further evidences on the optimum time for treatment and route of administration are required before effective translation of these findings into clinical practice. Bearing these in mind, this paper reviews the current literature discussing the involvement of miRNAs in major pathologies associated with ischaemic stroke and evaluates their value as reliable biomarkers and therapeutics for ischaemic stroke.
Collapse
Affiliation(s)
- Rais Reskiawan A Kadir
- Stroke, Division of Clinical Neuroscience, School of Medicine, The University of Nottingham, Clinical Sciences Building, Hucknall Road, Nottingham, NG5 1PB, UK
| | - Mansour Alwjwaj
- Stroke, Division of Clinical Neuroscience, School of Medicine, The University of Nottingham, Clinical Sciences Building, Hucknall Road, Nottingham, NG5 1PB, UK
| | - Ulvi Bayraktutan
- Stroke, Division of Clinical Neuroscience, School of Medicine, The University of Nottingham, Clinical Sciences Building, Hucknall Road, Nottingham, NG5 1PB, UK.
| |
Collapse
|
6
|
Gene-independent therapeutic interventions to maintain and restore light sensitivity in degenerating photoreceptors. Prog Retin Eye Res 2022; 90:101065. [PMID: 35562270 DOI: 10.1016/j.preteyeres.2022.101065] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 04/08/2022] [Accepted: 04/18/2022] [Indexed: 12/14/2022]
Abstract
Neurodegenerative retinal diseases are a prime cause of blindness in industrialized countries. In many cases, there are no therapeutic treatments, although they are essential to improve patients' quality of life. A set of disease-causing genes, which primarily affect photoreceptors, has already been identified and is of major interest for developing gene therapies. Nevertheless, depending on the nature and the state of the disease, gene-independent strategies are needed. Various strategies to halt disease progression or maintain function of the retina are under research. These therapeutic interventions include neuroprotection, direct reprogramming of affected photoreceptors, the application of non-coding RNAs, the generation of artificial photoreceptors by optogenetics and cell replacement strategies. During recent years, major breakthroughs have been made such as the first optogenetic application to a blind patient whose visual function partially recovered by targeting retinal ganglion cells. Also, RPE cell transplantation therapies are under clinical investigation and show great promise to improve visual function in blind patients. These cells are generated from human stem cells. Similar therapies for replacing photoreceptors are extensively tested in pre-clinical models. This marks just the start of promising new cures taking advantage of developments in the areas of genetic engineering, optogenetics, and stem-cell research. In this review, we present the recent therapeutic advances of gene-independent approaches that are currently under clinical evaluation. Our main focus is on photoreceptors as these sensory cells are highly vulnerable to degenerative diseases, and are crucial for light detection.
Collapse
|
7
|
Mead B, Tomarev S. The role of miRNA in retinal ganglion cell health and disease. Neural Regen Res 2022; 17:516-522. [PMID: 34380881 PMCID: PMC8504366 DOI: 10.4103/1673-5374.320974] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/19/2021] [Accepted: 04/15/2021] [Indexed: 12/03/2022] Open
Abstract
miRNA are short non-coding RNA responsible for the knockdown of proteins through their targeting and silencing of complimentary mRNA sequences. The miRNA landscape of a cell thus affects the levels of its proteins and has significant consequences to its health. Deviations in this miRNA landscape have been implicated in a variety of neurodegenerative diseases and have also garnered interest as targets for treatment. Retinal ganglion cells are the sole projection neuron of the retina with their axons making up the optic nerve. They are a focus of study not only for their importance in vision and the myriad of blinding diseases characterized by their dysfunction and loss, but also as a model of other central nervous system diseases such as spinal cord injury and traumatic brain injury. This review summarizes current knowledge on the role of miRNA in retinal ganglion cell function, highlighting how perturbations can result in disease, and how modulating their abundance may provide a novel avenue of therapeutic research.
Collapse
Affiliation(s)
- Ben Mead
- School of Optometry and Vision Sciences, Cardiff University, Cardiff, UK
| | - Stanislav Tomarev
- Section of Retinal Ganglion Cell Biology, Laboratory of Retinal Cell and Molecular Biology, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
8
|
Rajool Dezfuly A, Safaee A, Salehi H. Therapeutic effects of mesenchymal stem cells-derived extracellular vesicles' miRNAs on retinal regeneration: a review. Stem Cell Res Ther 2021; 12:530. [PMID: 34620234 PMCID: PMC8499475 DOI: 10.1186/s13287-021-02588-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 09/09/2021] [Indexed: 02/07/2023] Open
Abstract
Extracellular vesicles (EVs), which consist of microvesicles and exosomes, are secreted from all cells to transform vital information in the form of lipids, proteins, mRNAs and small RNAs such as microRNAs (miRNAs). Many studies demonstrated that EVs' miRNAs have effects on target cells. Numerous people suffer from the blindness caused by retinal degenerations. The death of retinal neurons is irreversible and creates permanent damage to the retina. In the absence of acceptable cures for retinal degenerative diseases, stem cells and their paracrine agents including EVs have become a promising therapeutic approach. Several studies showed that the therapeutic effects of stem cells are due to the miRNAs of their EVs. Considering the effects of microRNAs in retinal cells development and function and studies which provide the possible roles of mesenchymal stem cells-derived EVs miRNA content on retinal diseases, we focused on the similarities between these two groups of miRNAs that could be helpful for promoting new therapeutic techniques for retinal degenerative diseases.
Collapse
Affiliation(s)
- Ali Rajool Dezfuly
- Department of Anatomical and Molecular Biology Sciences, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Azadeh Safaee
- Department of Anatomical and Molecular Biology Sciences, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hossein Salehi
- Department of Anatomical and Molecular Biology Sciences, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
9
|
Kuznetsova AV, Rzhanova LA, Aleksandrova MA. Small Noncoding RNA in Regulation of Differentiation of Retinal Pigment Epithelium. Russ J Dev Biol 2021. [DOI: 10.1134/s106236042103005x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
10
|
Zhang D, Wu J, Wu J, Zhang S. Paeonol Induces Protective Autophagy in Retinal Photoreceptor Cells. Front Pharmacol 2021; 12:667959. [PMID: 34122088 PMCID: PMC8188238 DOI: 10.3389/fphar.2021.667959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 04/30/2021] [Indexed: 12/09/2022] Open
Abstract
Background: Retinal photoreceptor (RP) cells are widely involved in retina-related diseases, and oxidative stress plays a critical role in retinal secondary damage. Herein, we investigated the effectiveness and potential mechanisms of autophagy of paeonol (Pae) in terms of oxidation resistance. Methods: The animal model was induced by light damage (LD) in vivo, whereas the in vitro model was established by H2O2 stimulation. The effectiveness of Pae was evaluated by hematoxylin and eosin, terminal deoxynucleotidyl transferase dUTP nick end labeling assay, immunofluorescence, transmission electron microscopy, electroretinogram, and Western blot analysis in vivo, and the underlying mechanisms of Pae were assessed by Cell Counting Kit-8 assay, reactive oxygen species (ROS) assay, and Western blot analysis in 661W cells. We mainly evaluated the effects of Pae on apoptosis and autophagy. Results: Increased apoptosis of the LD-induced and decreased autophagy of RPs were mitigated by Pae treatment. Pea, which increased the expression of mitochondrial functional protein cytochrome c, reversed the decreased cell viability and autophagy induced by oxidative stress in 661W cells. Experiments showed that autophagy was downregulated in PINK1/Parkin dependent and the BNIP3L/Nix dependent pathways under H2O2 stimulation and was upregulated by Pae treatment. Pae increased the cell viability and reduced ROS levels through autophagy. Conclusion: Pretreatment with Pae preserved RP cells by enhancing autophagy, which protected retinal function.
Collapse
Affiliation(s)
- Daowei Zhang
- Eye Institute, Eye and ENT Hospital, College of Medicine, Fudan University, Shanghai, China
| | - Jiawen Wu
- Eye Institute, Eye and ENT Hospital, College of Medicine, Fudan University, Shanghai, China
| | - Jihong Wu
- Eye Institute, Eye and ENT Hospital, College of Medicine, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Science and Technology Commission of Shanghai Municipality, Shanghai, China.,State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Shanghai, China.,Key Laboratory of Myopia, Ministry of Health, Shanghai, China
| | - Shenghai Zhang
- Eye Institute, Eye and ENT Hospital, College of Medicine, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Science and Technology Commission of Shanghai Municipality, Shanghai, China.,State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Shanghai, China.,Key Laboratory of Myopia, Ministry of Health, Shanghai, China
| |
Collapse
|
11
|
Pawlick JS, Zuzic M, Pasquini G, Swiersy A, Busskamp V. MiRNA Regulatory Functions in Photoreceptors. Front Cell Dev Biol 2021; 8:620249. [PMID: 33553155 PMCID: PMC7858257 DOI: 10.3389/fcell.2020.620249] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 12/31/2020] [Indexed: 12/30/2022] Open
Abstract
MicroRNAs (miRNAs) are important regulators of gene expression. These small, non-coding RNAs post-transcriptionally silence messenger RNAs (mRNAs) in a sequence-specific manner. In this way, miRNAs control important regulatory functions, also in the retina. If dysregulated, these molecules are involved in several retinal pathologies. For example, several miRNAs have been linked to essential photoreceptor functions, including light sensitivity, synaptic transmission, and modulation of inflammatory responses. Mechanistic miRNA knockout and knockdown studies further linked their functions to degenerative retinal diseases. Of note, the type and timing of genetic manipulation before, during, or after retinal development, is important when studying specific miRNA knockout effects. Within this review, we focus on miR-124 and the miR-183/96/182 cluster, which have assigned functions in photoreceptors in health and disease. As a single miRNA can regulate hundreds of mRNAs, we will also discuss the experimental validation and manipulation approaches to study complex miRNA/mRNA regulatory networks. Revealing these networks is essential to understand retinal pathologies and to harness miRNAs as precise therapeutic and diagnostic tools to stabilize the photoreceptors’ transcriptomes and, thereby, function.
Collapse
Affiliation(s)
- Julia Sophie Pawlick
- Universitäts-Augenklinik Bonn, Department of Ophthalmology, University of Bonn, Bonn, Germany
| | - Marta Zuzic
- Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
| | - Giovanni Pasquini
- Universitäts-Augenklinik Bonn, Department of Ophthalmology, University of Bonn, Bonn, Germany.,Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
| | - Anka Swiersy
- Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
| | - Volker Busskamp
- Universitäts-Augenklinik Bonn, Department of Ophthalmology, University of Bonn, Bonn, Germany.,Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
12
|
Intartaglia D, Giamundo G, Conte I. The Impact of miRNAs in Health and Disease of Retinal Pigment Epithelium. Front Cell Dev Biol 2021; 8:589985. [PMID: 33520981 PMCID: PMC7844312 DOI: 10.3389/fcell.2020.589985] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 12/17/2020] [Indexed: 12/17/2022] Open
Abstract
MicroRNAs (miRNAs), a class of non-coding RNAs, are essential key players in the control of biological processes in both physiological and pathological conditions. miRNAs play important roles in fine tuning the expression of many genes, which often have roles in common molecular networks. miRNA dysregulation thus renders cells vulnerable to aberrant fluctuations in genes, resulting in degenerative diseases. The retinal pigment epithelium (RPE) is a monolayer of polarized pigmented epithelial cells that resides between the light-sensitive photoreceptors (PR) and the choriocapillaris. The demanding physiological functions of RPE cells require precise gene regulation for the maintenance of retinal homeostasis under stress conditions and the preservation of vision. Thus far, our understanding of how miRNAs function in the homeostasis and maintenance of the RPE has been poorly addressed, and advancing our knowledge is central to harnessing their potential as therapeutic agents to counteract visual impairment. This review focuses on the emerging roles of miRNAs in the function and health of the RPE and on the future exploration of miRNA-based therapeutic approaches to counteract blinding diseases.
Collapse
Affiliation(s)
| | | | - Ivan Conte
- Telethon Institute of Genetics and Medicine, Naples, Italy
- Department of Biology, Polytechnic and Basic Sciences School, University of Naples Federico II, Naples, Italy
| |
Collapse
|
13
|
Hammond CL, Roztocil E, Gonzalez MO, Feldon SE, Woeller CF. MicroRNA-130a Is Elevated in Thyroid Eye Disease and Increases Lipid Accumulation in Fibroblasts Through the Suppression of AMPK. Invest Ophthalmol Vis Sci 2021; 62:29. [PMID: 33507228 PMCID: PMC7846950 DOI: 10.1167/iovs.62.1.29] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Purpose Thyroid eye disease (TED) is a condition that causes the tissue behind the eye to become inflamed and can result in excessive fatty tissue accumulation in the orbit. Two subpopulations of fibroblasts reside in the orbit: those that highly express Thy1 (Thy1+) and those with little or no Thy1 (Thy1–). Thy1– orbital fibroblasts (OFs) are more prone to lipid accumulation than Thy1+ OFs. The purpose of this study was to investigate the mechanisms whereby Thy1– OFs more readily accumulate lipid. Methods We screened Thy1+ and Thy1– OFs for differences in microRNA (miRNA) expression. The effects of increasing miR-130a levels in OFs was investigated by measuring lipid accumulation and visualizing lipid deposits. To determine if adenosine monophosphate-activated protein kinase (AMPK) is important for lipid accumulation, we performed small interfering RNA (siRNA)-mediated knockdown of AMPKβ1. We measured AMPK expression and activity using immunoblotting for AMPK and AMPK target proteins. Results We determined that miR-130a was upregulated in Thy1– OFs and that miR-130a targets two subunits of AMPK. Increasing miR-130a levels enhanced lipid accumulation and reduced expression of AMPKα and AMPKβ in OFs. Depletion of AMPK also increased lipid accumulation. Activation of AMPK using AICAR attenuated lipid accumulation and increased phosphorylation of acetyl-CoA carboxylase (ACC) in OFs. Conclusions These data suggest that when Thy1– OFs accumulate in TED, miR-130a levels increase, leading to a decrease in AMPK activity. Decreased AMPK activity promotes lipid accumulation in TED OFs, leading to excessive fatty tissue accumulation in the orbit.
Collapse
Affiliation(s)
- Christine L Hammond
- Flaum Eye Institute, University of Rochester, Rochester, New York, United States
| | - Elisa Roztocil
- Flaum Eye Institute, University of Rochester, Rochester, New York, United States
| | - Mithra O Gonzalez
- Flaum Eye Institute, University of Rochester, Rochester, New York, United States
| | - Steven E Feldon
- Flaum Eye Institute, University of Rochester, Rochester, New York, United States
| | - Collynn F Woeller
- Flaum Eye Institute, University of Rochester, Rochester, New York, United States
| |
Collapse
|
14
|
Chu-Tan JA, Natoli R. The potential for microRNA-based therapeutics in retinal disorders. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:419. [PMID: 32395463 PMCID: PMC7210186 DOI: 10.21037/atm.2020.03.57] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Joshua A Chu-Tan
- The John Curtin School of Medical Research, College of Health and Medicine, The Australian National University, Acton, Australia.,The Australian National University Medical School, College of Health and Medicine, Acton, Australia
| | - Riccardo Natoli
- The John Curtin School of Medical Research, College of Health and Medicine, The Australian National University, Acton, Australia.,The Australian National University Medical School, College of Health and Medicine, Acton, Australia
| |
Collapse
|
15
|
Amini-Farsani Z, Asgharzade S. The impact of miR-183/182/96 gene regulation on the maturation, survival, and function of photoreceptor cells in the retina. J Comp Neurol 2019; 528:1616-1625. [PMID: 31785157 DOI: 10.1002/cne.24833] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Revised: 11/27/2019] [Accepted: 11/27/2019] [Indexed: 12/31/2022]
Abstract
MicroRNAs (MiRNAs) play important roles in posttranscriptional processes to regulate gene expression. MiRNAs control various biological processes, such as growth, development, and differentiation. The continuous physiological function of photoreceptors and retinal pigment epithelium requires precise regulation to maintain their homeostasis and function; hence, these cells are highly susceptible to premature death in retinal degenerative disorders. MiRNAs are essential for the retinal cell maturation and function; the miR-183 cluster represents one of the most important regulatory factors for the photoreceptor cells. Various studies together with bioinformatics analyses have shown that many genes contributing to the differentiation pathway of photoreceptors are targets of the miR-183 cluster, and the miR-183 cluster dysregulation causes certain defects in the differentiation of the photoreceptors and other retinal neurons by influencing the expression of target genes. Misexpression of miR-183 cluster in the human retinal epithelial cells leads to the reprogramming and transformation of these cells to neuron- and photoreceptor-like cells, which are associated with the expression of neuron- and photoreceptor-specific markers in human retinal pigment epitheliums cells. The knockout of this cluster causes the destruction of the outer segment of the photoreceptors, which subsequently causes the cells to exhibit severe susceptibility to light and eventually degenerate. Hundreds of target genes in this family are likely to affect the development and maintenance of the retina. Identifying the genes that are regulated by the miRNA-183 cluster provides researchers with important insights into the complex development and regeneration mechanism of the retina and may offer a new way for maintaining and regenerating photoreceptor cells in neurodegenerative diseases.
Collapse
Affiliation(s)
- Zeinab Amini-Farsani
- Young Researchers and Elites Club, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Samira Asgharzade
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
16
|
Wu KC, Chen XJ, Jin GH, Wang XY, Yang DD, Li YP, Xiang L, Zhang BW, Zhou GH, Zhang CJ, Jin ZB. Deletion of miR-182 Leads to Retinal Dysfunction in Mice. Invest Ophthalmol Vis Sci 2019; 60:1265-1274. [PMID: 30924851 DOI: 10.1167/iovs.18-24166] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose MicroRNA-182 (miR-182) is abundantly expressed in mammalian retinas; however, the association between miR-182 and retinal function remains unclear. In this study, we explored whether miR-182 contributes to functional decline in retinas using a miR-182 depleted mouse. Methods Electroretinogram (ERG) amplitudes at different ages were measured in miR-182 knockout (KO) mice. The thickness and lamination of retinas were assessed using a color fundus camera and high-resolution optical coherence tomography. Expression levels of key photoreceptor-specific genes and the miR-183/96/182 cluster (miR-183C) were quantified using quantitative real-time PCR. RNA sequencing and light-induced damage were carried out to observe the changes in the retinal transcriptome and sensitivity to light damage in the miR-182 KO mice. Results The ERG recording reveals that the ERG response amplitude decreased both at early and later ages when compared with control littermates. The expression of some key photoreceptor-specific genes was down-regulated with deletion of miR-182 in retina. RNA sequencing indicated that some biological processes of visual system were affected, and the numbers of potential target genes of miR-182 were presented in the mouse retina using bioinformatics analysis. The miR-182 KO mice were characterized by progressively losing the outer segment after being treated with light-damage exposure. The thickness and lamination of retina as well as compensatory expression of miR-183C showed no apparent changes in retina of miR-182 KO mice under normal laboratory lighting condition. Conclusions Our findings provided new insights into the relationship between the miR-182 and retinal development and revealed that miR-182 may play a critical role in maintaining retinal function.
Collapse
Affiliation(s)
- Kun-Chao Wu
- Lab for Stem Cell & Retinal Regeneration, Institute of Stem Cell Research, State Key Laboratory of Ophthalmology, Optometry and Vision Science, National Center for International Research in Regenerative Medicine and Neurogenetics, Wenzhou Medical University, Wenzhou, China.,Division of Ophthalmic Genetics, The Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Xue-Jiao Chen
- Lab for Stem Cell & Retinal Regeneration, Institute of Stem Cell Research, State Key Laboratory of Ophthalmology, Optometry and Vision Science, National Center for International Research in Regenerative Medicine and Neurogenetics, Wenzhou Medical University, Wenzhou, China.,Division of Ophthalmic Genetics, The Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Guang-Hui Jin
- Lab for Stem Cell & Retinal Regeneration, Institute of Stem Cell Research, State Key Laboratory of Ophthalmology, Optometry and Vision Science, National Center for International Research in Regenerative Medicine and Neurogenetics, Wenzhou Medical University, Wenzhou, China.,Division of Ophthalmic Genetics, The Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Xiao-Yun Wang
- Lab for Stem Cell & Retinal Regeneration, Institute of Stem Cell Research, State Key Laboratory of Ophthalmology, Optometry and Vision Science, National Center for International Research in Regenerative Medicine and Neurogenetics, Wenzhou Medical University, Wenzhou, China.,Division of Ophthalmic Genetics, The Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Dan-Dan Yang
- Lab for Stem Cell & Retinal Regeneration, Institute of Stem Cell Research, State Key Laboratory of Ophthalmology, Optometry and Vision Science, National Center for International Research in Regenerative Medicine and Neurogenetics, Wenzhou Medical University, Wenzhou, China.,Division of Ophthalmic Genetics, The Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Yan-Ping Li
- Lab for Stem Cell & Retinal Regeneration, Institute of Stem Cell Research, State Key Laboratory of Ophthalmology, Optometry and Vision Science, National Center for International Research in Regenerative Medicine and Neurogenetics, Wenzhou Medical University, Wenzhou, China.,Division of Ophthalmic Genetics, The Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Lue Xiang
- Lab for Stem Cell & Retinal Regeneration, Institute of Stem Cell Research, State Key Laboratory of Ophthalmology, Optometry and Vision Science, National Center for International Research in Regenerative Medicine and Neurogenetics, Wenzhou Medical University, Wenzhou, China.,Division of Ophthalmic Genetics, The Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Bo-Wen Zhang
- Lab for Stem Cell & Retinal Regeneration, Institute of Stem Cell Research, State Key Laboratory of Ophthalmology, Optometry and Vision Science, National Center for International Research in Regenerative Medicine and Neurogenetics, Wenzhou Medical University, Wenzhou, China.,Division of Ophthalmic Genetics, The Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Gao-Hui Zhou
- Lab for Stem Cell & Retinal Regeneration, Institute of Stem Cell Research, State Key Laboratory of Ophthalmology, Optometry and Vision Science, National Center for International Research in Regenerative Medicine and Neurogenetics, Wenzhou Medical University, Wenzhou, China.,Division of Ophthalmic Genetics, The Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Chang-Jun Zhang
- Lab for Stem Cell & Retinal Regeneration, Institute of Stem Cell Research, State Key Laboratory of Ophthalmology, Optometry and Vision Science, National Center for International Research in Regenerative Medicine and Neurogenetics, Wenzhou Medical University, Wenzhou, China.,Division of Ophthalmic Genetics, The Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Zi-Bing Jin
- Lab for Stem Cell & Retinal Regeneration, Institute of Stem Cell Research, State Key Laboratory of Ophthalmology, Optometry and Vision Science, National Center for International Research in Regenerative Medicine and Neurogenetics, Wenzhou Medical University, Wenzhou, China.,Division of Ophthalmic Genetics, The Eye Hospital, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
17
|
Banks SA, Pierce ML, Soukup GA. Sensational MicroRNAs: Neurosensory Roles of the MicroRNA-183 Family. Mol Neurobiol 2019; 57:358-371. [DOI: 10.1007/s12035-019-01717-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 07/19/2019] [Indexed: 12/20/2022]
|
18
|
Muhammad F, Trivett A, Wang D, Lee DJ. Tissue-specific production of MicroRNA-155 inhibits melanocortin 5 receptor-dependent suppressor macrophages to promote experimental autoimmune uveitis. Eur J Immunol 2019; 49:2074-2082. [PMID: 31177529 DOI: 10.1002/eji.201848073] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 05/16/2019] [Accepted: 06/03/2019] [Indexed: 12/16/2022]
Abstract
Tissue-specific immune regulation is an important component of the immune response relevant to many areas of immunology. The focus of this study is on tissue-specific mechanisms that contribute to autoimmune uveitis. Precise gene regulation is necessary for the proper expression of an inflammatory or regulatory response. This precision gene regulation can be accomplished by microRNA at the level of the mRNA transcript. miR-155, in particular, has a complicated role in the immune response with positive and negative inflammatory effects. In this work, we identify a decrease in miR-155 in suppressor macrophages and further examine how tissue-specific production of miR-155 impacts experimental autoimmune uveitis. Importantly, we show that eliminating miR-155 expression by the target tissue before initiation reduces disease severity, but elimination of miR-155 after the onset of inflammation does not alter the course of disease. Additionally, expression of miR-155 by the target tissue before initiation is necessary for the induction of regulatory immunity that protects from further autoimmune disease, but not after the onset of inflammation. In summary, we find a MC5r-dependent decrease in miR-155 in postexperimental autoimmune uveitis APC, miR-155 production by the target tissue is necessary for the initiation of autoimmune uveitis, and may have a role in establishing protective regulatory immunity.
Collapse
Affiliation(s)
- Fauziyya Muhammad
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Anna Trivett
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick National Laboratory for Cancer Research (FNLCR), Frederick, MD
| | - Dawei Wang
- Department of Ophthalmology/Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Darren J Lee
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK.,Department of Ophthalmology/Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| |
Collapse
|
19
|
Retinal miRNA Functions in Health and Disease. Genes (Basel) 2019; 10:genes10050377. [PMID: 31108959 PMCID: PMC6562649 DOI: 10.3390/genes10050377] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 05/15/2019] [Accepted: 05/15/2019] [Indexed: 01/12/2023] Open
Abstract
The health and function of our visual system relies on accurate gene expression. While many genetic mutations are associated with visual impairment and blindness, we are just beginning to understand the complex interplay between gene regulation and retinal pathologies. MicroRNAs (miRNAs), a class of non-coding RNAs, are important regulators of gene expression that exert their function through post-transcriptional silencing of complementary mRNA targets. According to recent transcriptomic analyses, certain miRNA species are expressed in all retinal cell types, while others are cell type-specific. As miRNAs play important roles in homeostasis, cellular function, and survival of differentiated retinal cell types, their dysregulation is associated with retinal degenerative diseases. Thus, advancing our understanding of the genetic networks modulated by miRNAs is central to harnessing their potential as therapeutic agents to overcome visual impairment. In this review, we summarize the role of distinct miRNAs in specific retinal cell types, the current knowledge on their implication in inherited retinal disorders, and their potential as therapeutic agents.
Collapse
|
20
|
Aldunate EZ, Di Foggia V, Di Marco F, Hervas LA, Ribeiro JC, Holder DL, Patel A, Jannini TB, Thompson DA, Martinez-Barbera JP, Pearson RA, Ali RR, Sowden JC. Conditional Dicer1 depletion using Chrnb4-Cre leads to cone cell death and impaired photopic vision. Sci Rep 2019; 9:2314. [PMID: 30783126 PMCID: PMC6381178 DOI: 10.1038/s41598-018-38294-9] [Citation(s) in RCA: 6] [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: 10/30/2017] [Accepted: 12/03/2018] [Indexed: 12/16/2022] Open
Abstract
Irreversible photoreceptor cell death is a major cause of blindness in many retinal dystrophies. A better understanding of the molecular mechanisms underlying the progressive loss of photoreceptor cells remains therefore crucial. Abnormal expression of microRNAs (miRNAs) has been linked with the aetiology of a number of retinal dystrophies. However, their role during the degenerative process remains poorly understood. Loss of cone photoreceptors in the human macula has the greatest impact on sight as these cells provide high acuity vision. Using a Chrnb4-cre; Dicerflox/flox conditional knockout mouse (Dicer CKO) to delete Dicer1 from cone cells, we show that cone photoreceptor cells degenerate and die in the Dicer-deleted retina. Embryonic eye morphogenesis appeared normal in Dicer CKO mice. Cone photoreceptor abnormalities were apparent by 3 weeks of age, displaying either very short or absent outer segments. By 4 months 50% of cones were lost and cone function was impaired as assessed by electroretinography (ERG). RNAseq analysis of the Dicer CKO retina revealed altered expression of genes involved in the visual perception pathway. These data show that loss of Dicer1 leads to early-onset cone cell degeneration and suggest that Dicer1 is essential for cone photoreceptor survival and homeostasis.
Collapse
Affiliation(s)
- Eduardo Zabala Aldunate
- Stem Cells and Regenerative Medicine Section, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK
| | - Valentina Di Foggia
- Stem Cells and Regenerative Medicine Section, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK
| | - Fabiana Di Marco
- Stem Cells and Regenerative Medicine Section, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK
| | - Laura Abelleira Hervas
- UCL Institute of Ophthalmology, Department of Genetics, London, 11-43 Bath Street, London, EC1V 9EL, UK
| | - Joana Claudio Ribeiro
- UCL Institute of Ophthalmology, Department of Genetics, London, 11-43 Bath Street, London, EC1V 9EL, UK
| | - Daniel L Holder
- Stem Cells and Regenerative Medicine Section, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK
| | - Aara Patel
- Stem Cells and Regenerative Medicine Section, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK
| | - Tommaso B Jannini
- Stem Cells and Regenerative Medicine Section, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK
| | - Dorothy A Thompson
- Clinical and Academic Department of Ophthalmology Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Juan Pedro Martinez-Barbera
- Developmental Biology of Birth Defects Section, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK
| | - Rachael A Pearson
- UCL Institute of Ophthalmology, Department of Genetics, London, 11-43 Bath Street, London, EC1V 9EL, UK
| | - Robin R Ali
- UCL Institute of Ophthalmology, Department of Genetics, London, 11-43 Bath Street, London, EC1V 9EL, UK
| | - Jane C Sowden
- Stem Cells and Regenerative Medicine Section, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK.
| |
Collapse
|
21
|
Taylor SM, Giuffre E, Moseley P, Hitchcock PF. The MicroRNA, miR-18a, Regulates NeuroD and Photoreceptor Differentiation in the Retina of Zebrafish. Dev Neurobiol 2019; 79:202-219. [PMID: 30615274 PMCID: PMC6351175 DOI: 10.1002/dneu.22666] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 12/13/2018] [Accepted: 12/14/2018] [Indexed: 12/21/2022]
Abstract
During embryonic retinal development, six types of retinal neurons are generated from multipotent progenitors in a strict spatiotemporal pattern. This pattern requires cell cycle exit (i.e. neurogenesis) and differentiation to be precisely regulated in a lineage‐specific manner. In zebrafish, the bHLH transcription factor NeuroD governs photoreceptor genesis through Notch signaling but also governs photoreceptor differentiation though distinct mechanisms that are currently unknown. Also unknown are the mechanisms that regulate NeuroD and the spatiotemporal pattern of photoreceptor development. Members of the miR‐17‐92 microRNA cluster regulate CNS neurogenesis, and a member of this cluster, miR‐18a, is predicted to target neuroD mRNA. The purpose of this study was to determine if, in the developing zebrafish retina, miR‐18a regulates NeuroD and if it plays a role in photoreceptor development. Quantitative RT‐PCR showed that, of the three miR‐18 family members (miR‐18a, b, and c), miR‐18a expression most closely parallels neuroD expression. Morpholino oligonucleotides and CRISPR/Cas9 gene editing were used for miR‐18a loss‐of‐function (LOF) and both resulted in larvae with more mature photoreceptors at 70 hpf without affecting cell proliferation. Western blot showed that miR‐18a LOF increases NeuroD protein levels and in vitro dual luciferase assay showed that miR‐18a directly interacts with the 3′ UTR of neuroD. Finally, tgif1 mutants have increased miR‐18a expression, less NeuroD protein and fewer mature photoreceptors, and the photoreceptor deficiency is rescued by miR‐18a knockdown. Together, these results show that, independent of neurogenesis, miR‐18a regulates the timing of photoreceptor differentiation and indicate that this occurs through post‐transcriptional regulation of NeuroD.
Collapse
Affiliation(s)
- Scott M Taylor
- Department of Biology, University of West Florida, 11000 University Parkway, Pensacola, Florida, 32514
| | - Emily Giuffre
- Department of Biology, University of West Florida, 11000 University Parkway, Pensacola, Florida, 32514
| | - Patience Moseley
- Department of Biology, University of West Florida, 11000 University Parkway, Pensacola, Florida, 32514
| | - Peter F Hitchcock
- Ophthalmology and Visual Sciences, University of Michigan, W. K. Kellogg Eye Center, 1000 Wall Street, Ann Arbor, Michigan, 48105
| |
Collapse
|
22
|
DICER1 Syndrome: Characterization of the Ocular Phenotype in a Family-Based Cohort Study. Ophthalmology 2018; 126:296-304. [PMID: 30339877 DOI: 10.1016/j.ophtha.2018.09.038] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 09/22/2018] [Accepted: 09/25/2018] [Indexed: 12/27/2022] Open
Abstract
PURPOSE To characterize the ocular phenotype of DICER1 syndrome. DESIGN Prospective, single-center, case-control study. PARTICIPANTS One hundred three patients with an identified germline pathogenic DICER1 variant (DICER1 carriers) and 69 family control participants underwent clinical and ophthalmic examination at the National Institutes of Health between 2011 and 2016. METHODS All participants were evaluated with a comprehensive ophthalmic examination, including best-corrected visual acuity, slit-lamp biomicroscopy, and a dilated fundus examination. A subset of patients returned for a more detailed evaluation including spectral-domain OCT, color fundus photography, fundus autofluorescence imaging, visual field testing, full-field electroretinography, and genetic testing for inherited retinal degenerative diseases. MAIN OUTCOME MEASURES Visual acuity and examination findings. RESULTS Most DICER1 carriers (97%) maintained a visual acuity of 20/40 or better in both eyes. Twenty-three DICER1 carriers (22%) showed ocular abnormalities compared with 4 family controls (6%; P = 0.005). These abnormalities included retinal pigment abnormalities (n = 6 [5.8%]), increased cup-to-disc ratio (n = 5 [4.9%]), optic nerve abnormalities (n = 2 [1.9%]), epiretinal membrane (n = 2 [1.9%]), and drusen (n = 2 [1.9%]). Overall, we observed a significant difference (P = 0.03) in the rate of retinal abnormalities in DICER1 carriers (n = 11 [11%]) versus controls (n = 1 [1.5%]). One patient demonstrated an unexpected diagnosis of retinitis pigmentosa with a novel variant of unknown significance in PRPF31, and 1 showed optic nerve elevation in the setting of increased intracranial pressure (ICP) of unclear cause. Three patients (3%) demonstrated DICER1-related ciliary body medulloepithelioma (CBME), 2 of which were identified during routine examination, a higher rate than that reported previously. CONCLUSIONS Ophthalmologists should be aware of the ophthalmic manifestations of DICER1 syndrome, and individuals and families should be counseled on the potential signs and symptoms. We recommend that children with a germline pathogenic variant in DICER1, especially those younger than 10 years, undergo annual dilated ophthalmic examination, looking for evidence of CBME, signs of increased ICP, and perhaps changes in the retinal pigment epithelium.
Collapse
|
23
|
Jun S, Datta S, Wang L, Pegany R, Cano M, Handa JT. The impact of lipids, lipid oxidation, and inflammation on AMD, and the potential role of miRNAs on lipid metabolism in the RPE. Exp Eye Res 2018; 181:346-355. [PMID: 30292489 DOI: 10.1016/j.exer.2018.09.023] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 08/31/2018] [Accepted: 09/30/2018] [Indexed: 12/17/2022]
Abstract
The accumulation of lipids within drusen, the epidemiologic link of a high fat diet, and the identification of polymorphisms in genes involved in lipid metabolism that are associated with disease risk, have prompted interest in the role of lipid abnormalities in AMD. Despite intensive investigation, our understanding of how lipid abnormalities contribute to AMD development remains unclear. Lipid metabolism is tightly regulated, and its dysregulation can trigger excess lipid accumulation within the RPE and Bruch's membrane. The high oxidative stress environment of the macula can promote lipid oxidation, impairing their original function as well as producing oxidation-specific epitopes (OSE), which unless neutralized, can induce unwanted inflammation that additionally contributes to AMD progression. Considering the multiple layers of lipid metabolism and inflammation, and the ability to simultaneously target multiple pathways, microRNA (miRNAs) have emerged as important regulators of many age-related diseases including atherosclerosis and Alzheimer's disease. These diseases have similar etiologic characteristics such as lipid-rich deposits, oxidative stress, and inflammation with AMD, which suggests that miRNAs might influence lipid metabolism in AMD. In this review, we discuss the contribution of lipids to AMD pathobiology and introduce how miRNAs might affect lipid metabolism during lesion development. Establishing how miRNAs contribute to lipid accumulation in AMD will help to define the role of lipids in AMD, and open new treatment avenues for this enigmatic disease.
Collapse
Affiliation(s)
- Sujung Jun
- Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, MD, 21287, United States
| | - Sayantan Datta
- Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, MD, 21287, United States
| | - Lei Wang
- Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, MD, 21287, United States
| | - Roma Pegany
- Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, MD, 21287, United States
| | - Marisol Cano
- Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, MD, 21287, United States
| | - James T Handa
- Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, MD, 21287, United States.
| |
Collapse
|
24
|
Non-coding RNAs in retinal development and function. Hum Genet 2018; 138:957-971. [DOI: 10.1007/s00439-018-1931-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 08/24/2018] [Indexed: 12/12/2022]
|
25
|
Chu-Tan JA, Rutar M, Saxena K, Aggio-Bruce R, Essex RW, Valter K, Jiao H, Fernando N, Wooff Y, Madigan MC, Provis J, Natoli R. MicroRNA-124 Dysregulation is Associated With Retinal Inflammation and Photoreceptor Death in the Degenerating Retina. Invest Ophthalmol Vis Sci 2018; 59:4094-4105. [PMID: 30098196 PMCID: PMC11647551 DOI: 10.1167/iovs.18-24623] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 06/27/2018] [Indexed: 11/24/2022] Open
Abstract
Purpose We sought to determine the role and retinal cellular location of microRNA-124 (miR-124) in a neuroinflammatory model of retinal degeneration. Further, we explored the anti-inflammatory relationship of miR-124 with a predicted messenger RNA (mRNA) binding partner, chemokine (C-C motif) ligand 2 (Ccl2), which is crucially involved in inflammatory cell recruitment in the damaged retina. Methods Human AMD donor eyes and photo-oxidative damaged (PD) mice were labeled for miR-124 expression using in situ hybridization. PDGFRa-cre RFP mice were used for Müller cell isolation from whole retinas. MIO-M1 immortalized cells and rat primary Müller cells were used for in vitro analysis of miR-124 expression and its relationship with Ccl2. Therapeutic efficacy was tested with intravitreal administration of miR-124 mimic in mice, with electroretinography used to determine retinal function. IBA1 immunohistochemistry and photoreceptor row counts were used for assessment of inflammation and cell death. Results MiR-124 expression was correlated with progressive retinal damage, inflammation, and cell death in human AMD and PD mice. In addition, miR-124 expression was inversely correlated to Ccl2 expression in mice following PD. MiR-124 was localized to both neuronal-like photoreceptors and glial (Müller) cells in the retina, with a redistribution from neurons to glia occurring as a consequence of PD. Finally, intravitreal administration of miR-124 mimics decreased retinal inflammation and photoreceptor cell death, and improved retinal function. Conclusions This study has provided an understanding of the mechanism behind miR-124 in the degenerating retina and demonstrates the usefulness of miR-124 mimics for the modulation of retinal degenerations.
Collapse
Affiliation(s)
- Joshua A. Chu-Tan
- The John Curtin School of Medical Research, The Australian National University, Acton, Australia
| | - Matt Rutar
- The John Curtin School of Medical Research, The Australian National University, Acton, Australia
| | - Kartik Saxena
- The John Curtin School of Medical Research, The Australian National University, Acton, Australia
| | - Riemke Aggio-Bruce
- The John Curtin School of Medical Research, The Australian National University, Acton, Australia
| | - Rohan W. Essex
- Academic Unit of Ophthalmology, The Australian National University, Canberra, Australia
| | - Krisztina Valter
- The John Curtin School of Medical Research, The Australian National University, Acton, Australia
- The Australian National University Medical School, Acton, Australia
| | - Haihan Jiao
- The John Curtin School of Medical Research, The Australian National University, Acton, Australia
| | - Nilisha Fernando
- The John Curtin School of Medical Research, The Australian National University, Acton, Australia
| | - Yvette Wooff
- The John Curtin School of Medical Research, The Australian National University, Acton, Australia
| | - Michele C. Madigan
- Save Sight Institute, Discipline of Clinical Ophthalmology, The University of Sydney, Sydney, Australia
- School of Optometry and Vision Science, The University of New South Wales, Kensington, Australia
| | - Jan Provis
- The John Curtin School of Medical Research, The Australian National University, Acton, Australia
- The Australian National University Medical School, Acton, Australia
| | - Riccardo Natoli
- The John Curtin School of Medical Research, The Australian National University, Acton, Australia
- The Australian National University Medical School, Acton, Australia
| |
Collapse
|
26
|
Li J, Hui L, Kang Q, Li R. Down-regulation of microRNA-27b promotes retinal pigment epithelial cell proliferation and migration by targeting Nox2. Pathol Res Pract 2018; 214:925-933. [DOI: 10.1016/j.prp.2018.05.025] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 05/21/2018] [Accepted: 05/22/2018] [Indexed: 12/11/2022]
|
27
|
Abstract
The small RNA regulatory molecules called microRNAs (miRNAs) play key roles in the development of most organisms. The expression of many different miRNAs has been described in the developing and mature vertebrate retina. The ability of miRNAs to regulate a diversity of messenger RNA targets allows them to have effects on many different developmental processes, but the functions of only a few miRNAs have been documented to date. Developmental transitions between cell states appear to be particularly sensitive to miRNA loss of function, as evidenced by specific miRNA knockdowns or from global perturbations in miRNA levels (e.g., Dicer deletion). However, we are still in only the very early stages of understanding the range of cellular functions miRNAs control during development.
Collapse
Affiliation(s)
- Thomas A Reh
- Department of Biological Structure, University of Washington, Seattle, Washington 98195, USA;
| | - Robert Hindges
- Centre for Developmental Neurobiology, MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, United Kingdom;
| |
Collapse
|
28
|
c-Jun-mediated microRNA-302d-3p induces RPE dedifferentiation by targeting p21 Waf1/Cip1. Cell Death Dis 2018; 9:451. [PMID: 29670082 PMCID: PMC5906557 DOI: 10.1038/s41419-018-0481-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 03/01/2018] [Accepted: 03/09/2018] [Indexed: 11/08/2022]
Abstract
Dedifferentiation of retinal pigment epithelium (RPE) cells and choroidal neovascularization (CNV) contributes to the pathogenesis of age-related macular degeneration (AMD). MicroRNAs (miRNAs) have crucial roles in AMD onset and progression. We thus aim to investigate the effects of miRNAs on RPE dedifferentiation and endothelium cell (EC) behavior, and analyze its downstream pathways. We have previously identified miR-302d-3p as the most downregulated miRNA signature along with RPE differentiation. Herein, in vitro study supported that miR-302d-3p induces RPE dedifferentiation typified by reduction of RPE characteristic markers, interrupts its phagocytosis, and promotes its migration, proliferation, and cell-cycle progression. c-Jun was identified as a potential upstream transcript factor for MIR302D, which might modulate RPE function by regulating miR-302d-3p expression. P21Waf1/Cip1, a cyclin-dependent kinase inhibitor encoded by the CDKN1A gene, was identified as a downstream target of miR-302d-3p. Our data suggested that p21Waf1/Cip1 could promote RPE differentiation, and inhibit its proliferation, migration, and cell-cycle progression. We also demonstrated that miR-302d-3p suppresses RPE differentiation through directly targeting p21Waf1/Cip1. In addition, the miR-302d-3p/CDKN1A axis was also involved in regulating tube formation of ECs, indicating its potential involvement in CNV formation. Taken together, our study implies that miR-302d-3p, regulated by c-Jun, contributes to the pathogenesis of both atrophic and exudative AMD. MiR-302d-3p promotes RPE dedifferentiation, migration, proliferation and cell-cycle progression, inhibits RPE phagocytosis, and induces abnormal EC behavior by targeting p21Waf1/Cip1. Pharmacological miR-302d-3p inhibitors are prospective therapeutic options for prevention and treatment of AMD.
Collapse
|
29
|
Jiang C, Qin B, Liu G, Sun X, Shi H, Ding S, Liu Y, Zhu M, Chen X, Zhao C. MicroRNA-184 promotes differentiation of the retinal pigment epithelium by targeting the AKT2/mTOR signaling pathway. Oncotarget 2018; 7:52340-52353. [PMID: 27418134 PMCID: PMC5239556 DOI: 10.18632/oncotarget.10566] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Accepted: 06/30/2016] [Indexed: 12/13/2022] Open
Abstract
Dedifferentiation of retinal pigment epithelium (RPE) cells is a crucial contributing factor to the pathology of retinal degenerative diseases, including age-related macular degeneration (AMD). Herein, we aim to reveal the roles of microRNAs (miRNAs) in RPE dedifferentiation and seek for potential therapeutic targets. Based on the microarray data, miR-184 was sorted out as the most up-regulated signature along with the differentiation from human induced pluripotent stem cells (hiPSC) to RPE cells, suggesting its potential promotive role in RPE differentiation. In vitro study indicated that miR-184 insufficiency suppressed RPE differentiation, typified by reduction of RPE markers, and promoted cell proliferation and migration. The role of miR-184 in maintaining regular RPE function was further proved in zebrafish studies. We also noticed that miR-184 expression was reduced in the macular RPE-choroid from a donor with RPE dysfunction compared to a healthy control. We next demonstrated that RAC-beta serine/threonine-protein kinase (AKT2) was a direct target for miR-184. MiR-184 promoted RPE differentiation via suppression of AKT2/mammalian target of rapamycin (mTOR) signaling pathway. We also found that AKT2 was up-regulated in macular RPE-choroid of the donor with RPE dysfunction and dry AMD patients. Taken together, our findings suggest that miR-184 insufficiency is involved in the pathogenesis of dry AMD. MiR-184 promotes RPE differentiation via inhibiting the AKT2/mTOR signaling pathway. MiR-184 based supplementary therapeutics and mTOR blocker, like rapamycin, are prospective options for AMD treatment.
Collapse
Affiliation(s)
- Chao Jiang
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University and State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Bing Qin
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University and State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China.,Department of Ophthalmology, The First People's Hospital of Suqian, Suqian, China
| | - Guohua Liu
- Department of Ophthalmology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Xiantao Sun
- Department of Ophthalmology, Children's Hospital of Zhengzhou, Zhengzhou, China
| | - Houxia Shi
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University and State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Sijia Ding
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University and State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Yuan Liu
- Department of Ophthalmology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Meidong Zhu
- Save Sight Institute, Discipline of Clinical Ophthalmology and Eye Health (CO9), The University of Sydney, Sydney, Australia
| | - Xue Chen
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University and State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Chen Zhao
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University and State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China.,Department of Ophthalmology, Eye and ENT Hospital, Fudan University, Shanghai, China.,State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat Sen University, Guangzhou, China
| |
Collapse
|
30
|
Donato L, Bramanti P, Scimone C, Rinaldi C, Giorgianni F, Beranova-Giorgianni S, Koirala D, D'Angelo R, Sidoti A. miRNAexpression profile of retinal pigment epithelial cells under oxidative stress conditions. FEBS Open Bio 2018; 8:219-233. [PMID: 29435412 PMCID: PMC5794457 DOI: 10.1002/2211-5463.12360] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 11/03/2017] [Accepted: 11/24/2017] [Indexed: 12/22/2022] Open
Abstract
Deep analysis of regulative mechanisms of transcription and translation in eukaryotes could improve knowledge of many genetic pathologies such as retinitis pigmentosa (RP). New layers of complexity have recently emerged with the discovery that ‘junk’ DNA is transcribed and, among these, miRNAs have assumed a preponderant role. We compared changes in the expression of miRNAs obtained from whole transcriptome analyses, between two groups of retinal pigment epithelium (RPE) cells, one untreated and the other exposed to the oxidant agent oxidized low‐density lipoprotein (oxLDL), examining four time points (1, 2, 4 and 6 h). We found that 23 miRNAs exhibited altered expression in the treated samples, targeting genes involved in several biochemical pathways, many of them associated to RP for the first time, such as those mediated by insulin receptor signaling and son of sevenless. Moreover, five RP causative genes (KLHL7, RDH11,CERKL, AIPL1 and USH1G) emerged as already validated targets of five altered miRNAs (hsa‐miR‐1307, hsa‐miR‐3064, hsa‐miR‐4709, hsa‐miR‐3615 and hsa‐miR‐637), suggesting a tight connection between induced oxidative stress and RP development and progression. This miRNA expression analysis of oxidative stress‐induced RPE cells has discovered new regulative functions of miRNAs in RP that should lead to the discovery of new ways to regulate the etiopathogenesis of RP.
Collapse
Affiliation(s)
- Luigi Donato
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging Division of Medical Biotechnologies and Preventive Medicine University of Messina Italy.,Department of Cutting-Edge Medicine and Therapies Biomolecular Strategies and Neuroscience Section of Neuroscience-applied, Molecular Genetics and Predictive MedicineI.E.M E.S.T. Palermo Italy
| | | | - Concetta Scimone
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging Division of Medical Biotechnologies and Preventive Medicine University of Messina Italy.,Department of Cutting-Edge Medicine and Therapies Biomolecular Strategies and Neuroscience Section of Neuroscience-applied, Molecular Genetics and Predictive MedicineI.E.M E.S.T. Palermo Italy
| | - Carmela Rinaldi
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging Division of Medical Biotechnologies and Preventive Medicine University of Messina Italy
| | | | | | | | - Rosalia D'Angelo
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging Division of Medical Biotechnologies and Preventive Medicine University of Messina Italy
| | - Antonina Sidoti
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging Division of Medical Biotechnologies and Preventive Medicine University of Messina Italy.,Department of Cutting-Edge Medicine and Therapies Biomolecular Strategies and Neuroscience Section of Neuroscience-applied, Molecular Genetics and Predictive MedicineI.E.M E.S.T. Palermo Italy
| |
Collapse
|
31
|
Wohl SG, Jorstad NL, Levine EM, Reh TA. Müller glial microRNAs are required for the maintenance of glial homeostasis and retinal architecture. Nat Commun 2017; 8:1603. [PMID: 29150673 PMCID: PMC5693933 DOI: 10.1038/s41467-017-01624-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 10/02/2017] [Indexed: 01/21/2023] Open
Abstract
To better understand the roles of microRNAs in glial function, we used a conditional deletion of Dicer1 (Dicer-CKOMG) in retinal Müller glia (MG). Dicer1 deletion from the MG leads to an abnormal migration of the cells as early as 1 month after the deletion. By 6 months after Dicer1 deletion, the MG form large aggregations and severely disrupt normal retinal architecture and function. The most highly upregulated gene in the Dicer-CKOMG MG is the proteoglycan Brevican (Bcan) and overexpression of Bcan results in similar aggregations of the MG in wild-type retina. One potential microRNA that regulates Bcan is miR-9, and overexpression of miR-9 can partly rescue the effects of Dicer1 deletion on the MG phenotype. We also find that MG from retinitis pigmentosa patients display an increase in Brevican immunoreactivity at sites of MG aggregation, linking the retinal remodeling that occurs in chronic disease with microRNAs.
Collapse
Affiliation(s)
- Stefanie G Wohl
- Department of Biological Structure, University of Washington, Health Sciences Center, Box 357420, 1959 Pacific Street NE, Seattle, WA, 98195, USA
| | - Nikolas L Jorstad
- Department of Biological Structure, University of Washington, Health Sciences Center, Box 357420, 1959 Pacific Street NE, Seattle, WA, 98195, USA
| | - Edward M Levine
- Department of Ophthalmology and Visual Sciences, Vanderbilt University, Nashville, TN, 37232, USA
| | - Thomas A Reh
- Department of Biological Structure, University of Washington, Health Sciences Center, Box 357420, 1959 Pacific Street NE, Seattle, WA, 98195, USA.
| |
Collapse
|
32
|
Quintero H, Lamas M. microRNA expression in the neural retina: Focus on Müller glia. J Neurosci Res 2017; 96:362-370. [DOI: 10.1002/jnr.24181] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 08/18/2017] [Accepted: 08/23/2017] [Indexed: 12/16/2022]
Affiliation(s)
- Heberto Quintero
- Departamento de Farmacobiología; Cinvestav Sede Sur; Mexico City Mexico
- Department of Neuroscience; University of Montreal Hospital Research Centre (CRCHUM); Montreal Quebec Canada
| | - Mónica Lamas
- Departamento de Farmacobiología; Cinvestav Sede Sur; Mexico City Mexico
| |
Collapse
|
33
|
Sundermeier TR, Sakami S, Sahu B, Howell SJ, Gao S, Dong Z, Golczak M, Maeda A, Palczewski K. MicroRNA-processing Enzymes Are Essential for Survival and Function of Mature Retinal Pigmented Epithelial Cells in Mice. J Biol Chem 2017; 292:3366-3378. [PMID: 28104803 DOI: 10.1074/jbc.m116.770024] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 01/17/2017] [Indexed: 11/06/2022] Open
Abstract
Age-related macular degeneration (AMD) is a major cause of irreversible vision loss. The neovascular or "wet" form of AMD can be treated to varying degrees with anti-angiogenic drugs, but geographic atrophy (GA) is an advanced stage of the more prevalent "dry" form of AMD for which there is no effective treatment. Development of GA has been linked to loss of the microRNA (miRNA)-processing enzyme DICER1 in the mature retinal pigmented epithelium (RPE). This loss results in the accumulation of toxic transcripts of Alu transposable elements, which activate the NLRP3 inflammasome and additional downstream pathways that compromise the integrity and function of the RPE. However, it remains unclear whether the loss of miRNA processing and subsequent gene regulation in the RPE due to DICER1 deficiency also contributes to RPE cell death. To clarify the role of miRNAs in RPE cells, we used two different mature RPE cell-specific Cre recombinase drivers to inactivate either Dicer1 or DiGeorge syndrome critical region 8 (Dgcr8), thus removing RPE miRNA regulatory activity in mice by disrupting two independent and essential steps of miRNA biogenesis. In contrast with prior studies, we found that the loss of each factor independently led to strikingly similar defects in the survival and function of the RPE and retina. These results suggest that the loss of miRNAs also contributes to RPE cell death and loss of visual function and could affect the pathology of dry AMD.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Marcin Golczak
- Departments of Pharmacology; Cleveland Center for Membrane and Structural Biology, Case Western Reserve University, Cleveland, Ohio 44106
| | - Akiko Maeda
- Ophthalmology and Visual Sciences, School of Medicine
| | - Krzysztof Palczewski
- Departments of Pharmacology; Cleveland Center for Membrane and Structural Biology, Case Western Reserve University, Cleveland, Ohio 44106.
| |
Collapse
|
34
|
Dash S, Siddam AD, Barnum CE, Janga SC, Lachke SA. RNA-binding proteins in eye development and disease: implication of conserved RNA granule components. WILEY INTERDISCIPLINARY REVIEWS-RNA 2016; 7:527-57. [PMID: 27133484 DOI: 10.1002/wrna.1355] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 03/21/2016] [Indexed: 01/16/2023]
Abstract
The molecular biology of metazoan eye development is an area of intense investigation. These efforts have led to the surprising recognition that although insect and vertebrate eyes have dramatically different structures, the orthologs or family members of several conserved transcription and signaling regulators such as Pax6, Six3, Prox1, and Bmp4 are commonly required for their development. In contrast, our understanding of posttranscriptional regulation in eye development and disease, particularly regarding the function of RNA-binding proteins (RBPs), is limited. We examine the present knowledge of RBPs in eye development in the insect model Drosophila as well as several vertebrate models such as fish, frog, chicken, and mouse. Interestingly, of the 42 RBPs that have been investigated for their expression or function in vertebrate eye development, 24 (~60%) are recognized in eukaryotic cells as components of RNA granules such as processing bodies, stress granules, or other specialized ribonucleoprotein (RNP) complexes. We discuss the distinct developmental and cellular events that may necessitate potential RBP/RNA granule-associated RNA regulon models to facilitate posttranscriptional control of gene expression in eye morphogenesis. In support of these hypotheses, three RBPs and RNP/RNA granule components Tdrd7, Caprin2, and Stau2 are linked to ocular developmental defects such as congenital cataract, Peters anomaly, and microphthalmia in human patients or animal models. We conclude by discussing the utility of interdisciplinary approaches such as the bioinformatics tool iSyTE (integrated Systems Tool for Eye gene discovery) to prioritize RBPs for deriving posttranscriptional regulatory networks in eye development and disease. WIREs RNA 2016, 7:527-557. doi: 10.1002/wrna.1355 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Soma Dash
- Department of Biological Sciences, University of Delaware, Newark, DE, USA
| | - Archana D Siddam
- Department of Biological Sciences, University of Delaware, Newark, DE, USA
| | - Carrie E Barnum
- Department of Biological Sciences, University of Delaware, Newark, DE, USA
| | - Sarath Chandra Janga
- Department of Biohealth Informatics, School of Informatics and Computing, Indiana University & Purdue University Indianapolis, Indianapolis, IN, USA.,Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Salil A Lachke
- Department of Biological Sciences, University of Delaware, Newark, DE, USA.,Center for Bioinformatics and Computational Biology, University of Delaware, Newark, DE, USA
| |
Collapse
|
35
|
Svobodova E, Kubikova J, Svoboda P. Production of small RNAs by mammalian Dicer. Pflugers Arch 2016; 468:1089-102. [PMID: 27048428 PMCID: PMC4893058 DOI: 10.1007/s00424-016-1817-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Revised: 03/20/2016] [Accepted: 03/24/2016] [Indexed: 01/16/2023]
Abstract
MicroRNA (miRNA) and RNA interference (RNAi) pathways employ RNase III Dicer for the biogenesis of small RNAs guiding post-transcriptional repression. Requirements for Dicer activity differ in the two pathways. The biogenesis of miRNAs requires a single Dicer cleavage of a short hairpin precursor to produce a small RNA with a precisely defined sequence, while small RNAs in RNAi come from a processive cleavage of a long double-stranded RNA (dsRNA) into a pool of small RNAs with different sequences. While Dicer is generally conserved among eukaryotes, its substrate recognition, cleavage, and biological roles differ. In Metazoa, a single Dicer can function as a universal factor for RNAi and miRNA pathways or as a factor adapted specifically for one of the pathways. In this review, we focus on the structure, function, and evolution of mammalian Dicer. We discuss key structural features of Dicer and other factors defining Dicer substrate repertoire and biological functions in mammals in comparison with invertebrate models. The key for adaptation of Dicer for miRNA or RNAi pathways is the N-terminal helicase, a dynamically evolving Dicer domain. Its functionality differs between mammals and invertebrates: the mammalian Dicer is well adapted to produce miRNAs while its ability to support RNAi is limited.
Collapse
Affiliation(s)
- Eliska Svobodova
- Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Videnska 1083, Prague 4, 142 20, Czech Republic
| | - Jana Kubikova
- Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Videnska 1083, Prague 4, 142 20, Czech Republic
| | - Petr Svoboda
- Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Videnska 1083, Prague 4, 142 20, Czech Republic.
| |
Collapse
|
36
|
microRNA-34a-Mediated Down-Regulation of the Microglial-Enriched Triggering Receptor and Phagocytosis-Sensor TREM2 in Age-Related Macular Degeneration. PLoS One 2016; 11:e0150211. [PMID: 26949937 PMCID: PMC4780721 DOI: 10.1371/journal.pone.0150211] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 02/10/2016] [Indexed: 12/17/2022] Open
Abstract
The aggregation of Aβ42-peptides and the formation of drusen in age-related macular degeneration (AMD) are due in part to the inability of homeostatic phagocytic mechanisms to clear self-aggregating Aβ42-peptides from the extracellular space. The triggering receptor expressed in myeloid/microglial cells-2 (TREM2), a trans-membrane-spanning, sensor-receptor of the immune-globulin/lectin-like gene superfamily is a critical component of Aβ42-peptide clearance. Here we report a significant deficit in TREM2 in AMD retina and in cytokine- or oxidatively-stressed microglial (MG) cells. RT-PCR, miRNA-array, LED-Northern and Western blot studies indicated up-regulation of a microglial-enriched NF-кB-sensitive miRNA-34a coupled to a down-regulation of TREM2 in the same samples. Bioinformatics/transfection-luciferase reporter assays indicated that miRNA-34a targets the 299 nucleotide TREM2-mRNA-3'UTR, resulting in TREM2 down-regulation. C8B4-microglial cells challenged with Aβ42 were able to phagocytose these peptides, while miRNA-34a down-regulated both TREM2 and the ability of microglial-cells to phagocytose. Treatment of TNFα-stressed MG cells with phenyl-butyl nitrone (PBN), caffeic-acid phenethyl ester (CAPE), the NF-kB - [corrected] inhibitor/resveratrol analog CAY10512 or curcumin abrogated these responses. Incubation of anti-miRNA-34a (AM-34a) normalized miRNA-34a abundance and restored TREM2 back to homeostatic levels. These data support five novel observations: (i) that a ROS- and NF-kB - [corrected] sensitive, miRNA-34a-mediated modulation of TREM2 may in part regulate the phagocytic response; (ii) that gene products encoded on two different chromosomes (miRNA-34a at chr1q36.22 and TREM2 at chr6p21.1) orchestrate a phagocytic-Aβ42-peptide clearance-system; (iii) that this NF-kB-mediated-miRNA-34a-TREM2 mechanism is inducible from outside of the cell; (iv) that when operating normally, this pathway can clear Aβ42 peptide monomers from the extracellular medium; and (v) that anti-NF-kB and/or anti-miRNA (AM)-based therapeutic strategies may be useful against deficits in TREM-2 receptor-based-sensing and -phagocytic signaling that promote pathogenic amyloidogenesis.
Collapse
|