1
|
Li P, Yu H, Zhang G, Kang L, Qin B, Cao Y, Luo J, Chen X, Wang Y, Qin M, Wu J, Huang Y, Zou X, Guan H, Wang Y. Identification and Characterization of N6-Methyladenosine CircRNAs and Methyltransferases in the Lens Epithelium Cells From Age-Related Cataract. Invest Ophthalmol Vis Sci 2021; 61:13. [PMID: 32761139 PMCID: PMC7441297 DOI: 10.1167/iovs.61.10.13] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Purpose To explore the involvement of N6-methyladenosine (m6A) modification in circular RNAs (circRNAs) and relevant methyltransferases in the lesion of lens epithelium cells (LECs) under the circumstances of age-related cataract (ARC). Methods LECs were collected from normal subjects and patients with cortical type of ARC (ARCC). M6A-tagged circRNAs and circRNAs expression were analyzed by m6A-modified RNA immunoprecipitation sequencing (m6A-RIP-seq) and RNA sequencing (RNA-seq). Gene Ontology (GO) annotation and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were used to predict possible functions of the m6A-circRNAs. Expression of m6A-related methyltransferase and demethytransferase was measured by quantitative real-time polymerase chain reaction. Expression and location of AlkB homolog 5 RNA demethylase (ALKBH5), a key component of m6A demethytransferase, were determined by Western blot and immunostaining. Results All 4646 m6A peaks within circRNAs had different abundances, with 2472 enriched and 2174 subdued. The level of m6A abundance in total circRNAs was decreased in the LECs from ARCCs in comparison with the controls. We also found that the expression of highly m6A-tagged circRNAs was mostly decreased in comparison with non-m6A-tagged circRNAs. The bioinformatics analysis predicted the potential functions of m6A modified circRNAs and the relevant pathways that may be associated with m6A modified circRNAs. Among five major methyltransferases, ALKBH5 was significantly upregulated in LECs of ARCCs. Conclusions Our data provided novel evidence regarding the involvement of circRNAs m6A modifications in ARC. The altered expression of methyltransferases in lens tissue might selectively change the epigenetic profile of lens genome through regulating genes that host the circRNAs, thus enhance the susceptibility to ARC. The results might provide a new insight in the molecular target of ARC pathogenesis.
Collapse
|
2
|
Loera-Valencia R, Cedazo-Minguez A, Kenigsberg PA, Page G, Duarte AI, Giusti P, Zusso M, Robert P, Frisoni GB, Cattaneo A, Zille M, Boltze J, Cartier N, Buee L, Johansson G, Winblad B. Current and emerging avenues for Alzheimer's disease drug targets. J Intern Med 2019; 286:398-437. [PMID: 31286586 DOI: 10.1111/joim.12959] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Alzheimer's disease (AD), the most frequent cause of dementia, is escalating as a global epidemic, and so far, there is neither cure nor treatment to alter its progression. The most important feature of the disease is neuronal death and loss of cognitive functions, caused probably from several pathological processes in the brain. The main neuropathological features of AD are widely described as amyloid beta (Aβ) plaques and neurofibrillary tangles of the aggregated protein tau, which contribute to the disease. Nevertheless, AD brains suffer from a variety of alterations in function, such as energy metabolism, inflammation and synaptic activity. The latest decades have seen an explosion of genes and molecules that can be employed as targets aiming to improve brain physiology, which can result in preventive strategies for AD. Moreover, therapeutics using these targets can help AD brains to sustain function during the development of AD pathology. Here, we review broadly recent information for potential targets that can modify AD through diverse pharmacological and nonpharmacological approaches including gene therapy. We propose that AD could be tackled not only using combination therapies including Aβ and tau, but also considering insulin and cholesterol metabolism, vascular function, synaptic plasticity, epigenetics, neurovascular junction and blood-brain barrier targets that have been studied recently. We also make a case for the role of gut microbiota in AD. Our hope is to promote the continuing research of diverse targets affecting AD and promote diverse targeting as a near-future strategy.
Collapse
Affiliation(s)
- R Loera-Valencia
- Division of Neurogeriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
| | - A Cedazo-Minguez
- Division of Neurogeriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
| | | | - G Page
- Neurovascular Unit and Cognitive impairments - EA3808, University of Poitiers, Poitiers, France
| | - A I Duarte
- CNC- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal
| | - P Giusti
- Dipartimento di Scienze del Farmaco, Università degli Studi di Padova, Padova, Italy
| | - M Zusso
- Dipartimento di Scienze del Farmaco, Università degli Studi di Padova, Padova, Italy
| | - P Robert
- CoBTeK - lab, CHU Nice University Côte d'Azur, Nice, France
| | - G B Frisoni
- University Hospitals and University of Geneva, Geneva, Switzerland
| | - A Cattaneo
- University Hospitals and University of Geneva, Geneva, Switzerland
| | - M Zille
- Institute of Experimental and Clinical Pharmacology and Toxicology, Lübeck, Germany
| | - J Boltze
- School of Life Sciences, The University of Warwick, Coventry, UK
| | - N Cartier
- Preclinical research platform, INSERM U1169/MIRCen Commissariat à l'énergie atomique, Fontenay aux Roses, France.,Université Paris-Sud, Orsay, France
| | - L Buee
- Alzheimer & Tauopathies, LabEx DISTALZ, CHU-Lille, Inserm, Univ. Lille, Lille, France
| | - G Johansson
- Division of Neurogeriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
| | - B Winblad
- Division of Neurogeriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden.,Theme Aging, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
3
|
Zusso M, Barbierato M, Facci L, Skaper SD, Giusti P. Neuroepigenetics and Alzheimer's Disease: An Update. J Alzheimers Dis 2019; 64:671-688. [PMID: 29991138 DOI: 10.3233/jad-180259] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Epigenetics is the study of changes in gene expression which may be triggered by both genetic and environmental factors, and independent from changes to the underlying DNA sequence-a change in phenotype without a change in genotype-which in turn affects how cells read genes. Epigenetic changes represent a regular and natural occurrence but can be influenced also by factors such as age, environment, and disease state. Epigenetic modifications can manifest themselves not only as the manner in which cells terminally differentiate, but can have also deleterious effects, resulting in diseases such as cancer. At least three systems including DNA methylation, histone modification, and non-coding RNA (ncRNA)-associated gene silencing are thought to initiate and sustain epigenetic change. For example, in Alzheimer's disease (AD), both genetic and non-genetic factors contribute to disease etiopathology. While over 250 gene mutations have been related to familial AD, less than 5% of AD cases are explained by known disease genes. More than likely, non-genetic factors, probably triggered by environmental factors, are causative factors of late-onset AD. AD is associated with dysregulation of DNA methylation, histone modifications, and ncRNAs. Among the classes of ncRNA, microRNAs (miRNAs) have a well-established regulatory relevance. MicroRNAs are highly expressed in CNS neurons, where they play a major role in neuron differentiation, synaptogenesis, and plasticity. MicroRNAs impact higher cognitive functions, as their functional impairment is involved in the etiology of neurological diseases, including AD. Alterations in the miRNA network contribute to AD disease processes, e.g., in the regulation of amyloid peptides, tau, lipid metabolism, and neuroinflammation. MicroRNAs, both as biomarkers for AD and therapeutic targets, are in the early stages of exploration. In addition, emerging data suggest that altered transcription of long ncRNAs, endogenous, ncRNAs longer than 200 nucleotides, may be involved in an elevated risk for AD.
Collapse
Affiliation(s)
- Morena Zusso
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Largo Meneghetti, Padua, Italy
| | - Massimo Barbierato
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Largo Meneghetti, Padua, Italy
| | - Laura Facci
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Largo Meneghetti, Padua, Italy
| | - Stephen D Skaper
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Largo Meneghetti, Padua, Italy
| | - Pietro Giusti
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Largo Meneghetti, Padua, Italy
| |
Collapse
|
4
|
Neal M, Richardson JR. Epigenetic regulation of astrocyte function in neuroinflammation and neurodegeneration. Biochim Biophys Acta Mol Basis Dis 2017; 1864:432-443. [PMID: 29113750 DOI: 10.1016/j.bbadis.2017.11.004] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 10/22/2017] [Accepted: 11/02/2017] [Indexed: 01/01/2023]
Abstract
Epigenetic mechanisms control various functions throughout the body, from cell fate determination in development to immune responses and inflammation. Neuroinflammation is one of the prime contributors to the initiation and progression of neurodegeneration in a variety of diseases, including Alzheimer's and Parkinson's diseases. Because astrocytes are the largest population of glial cells, they represent an important regulator of CNS function, both in health and disease. Only recently have studies begun to identify the epigenetic mechanisms regulating astrocyte responses in neurodegenerative diseases. These epigenetic mechanisms, along with the epigenetic marks involved in astrocyte development, could elucidate novel pathways to potentially modulate astrocyte-mediated neuroinflammation and neurotoxicity. This review examines the known epigenetic mechanisms involved in regulation of astrocyte function, from development to neurodegeneration, and links these mechanisms to potential astrocyte-specific roles in neurodegenerative disease with a focus on potential opportunities for therapeutic intervention.
Collapse
Affiliation(s)
- Matthew Neal
- Department of Pharmaceutical Sciences and Center for Neurodegenerative Disease and Aging, Northeast Ohio Medical University, Rootstown, OH 44201, USA
| | - Jason R Richardson
- Department of Pharmaceutical Sciences and Center for Neurodegenerative Disease and Aging, Northeast Ohio Medical University, Rootstown, OH 44201, USA.
| |
Collapse
|
5
|
Pistollato F, Ohayon EL, Lam A, Langley GR, Novak TJ, Pamies D, Perry G, Trushina E, Williams RS, Roher AE, Hartung T, Harnad S, Barnard N, Morris MC, Lai MC, Merkley R, Chandrasekera PC. Alzheimer disease research in the 21st century: past and current failures, new perspectives and funding priorities. Oncotarget 2016; 7:38999-39016. [PMID: 27229915 PMCID: PMC5129909 DOI: 10.18632/oncotarget.9175] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 04/18/2016] [Indexed: 12/20/2022] Open
Abstract
Much of Alzheimer disease (AD) research has been traditionally based on the use of animals, which have been extensively applied in an effort to both improve our understanding of the pathophysiological mechanisms of the disease and to test novel therapeutic approaches. However, decades of such research have not effectively translated into substantial therapeutic success for human patients. Here we critically discuss these issues in order to determine how existing human-based methods can be applied to study AD pathology and develop novel therapeutics. These methods, which include patient-derived cells, computational analysis and models, together with large-scale epidemiological studies represent novel and exciting tools to enhance and forward AD research. In particular, these methods are helping advance AD research by contributing multifactorial and multidimensional perspectives, especially considering the crucial role played by lifestyle risk factors in the determination of AD risk. In addition to research techniques, we also consider related pitfalls and flaws in the current research funding system. Conversely, we identify encouraging new trends in research and government policy. In light of these new research directions, we provide recommendations regarding prioritization of research funding. The goal of this document is to stimulate scientific and public discussion on the need to explore new avenues in AD research, considering outcome and ethics as core principles to reliably judge traditional research efforts and eventually undertake new research strategies.
Collapse
Affiliation(s)
| | - Elan L. Ohayon
- Green Neuroscience Laboratory, Neurolinx Research Institute, San Diego, CA, USA
| | - Ann Lam
- Physicians Committee for Responsible Medicine, Washington, DC, USA
- Green Neuroscience Laboratory, Neurolinx Research Institute, San Diego, CA, USA
| | - Gillian R. Langley
- Research and Toxicology Department, Humane Society International, London, UK
| | | | - David Pamies
- CAAT, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - George Perry
- College of Sciences, University of Texas at San Antonio, San Antonio, TX, USA
| | | | - Robin S.B. Williams
- Centre for Biomedical Sciences, School of Biological Sciences, Royal Holloway University of London, Egham, UK
| | - Alex E. Roher
- Division of Clinical Education, Midwestern University, Glendale, AZ, USA
- Division of Neurobiology, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Thomas Hartung
- CAAT, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Stevan Harnad
- Department of Psychology, University of Quebec/Montreal, Montreal, Canada
| | - Neal Barnard
- Physicians Committee for Responsible Medicine, Washington, DC, USA
| | - Martha Clare Morris
- Section of Nutrition and Nutritional Epidemiology, Department of Internal Medicine, Rush University, Chicago, IL, USA
| | - Mei-Chun Lai
- Physicians Committee for Responsible Medicine, Washington, DC, USA
| | - Ryan Merkley
- Physicians Committee for Responsible Medicine, Washington, DC, USA
| | | |
Collapse
|
6
|
Lundstrom K. Personalized Medicine and Epigenetic Drug Development. PERSONALIZED EPIGENETICS 2015:369-386. [DOI: 10.1016/b978-0-12-420135-4.00013-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
7
|
Satterlee JS, Beckel-Mitchener A, Little R, Procaccini D, Rutter JL, Lossie AC. Neuroepigenomics: Resources, Obstacles, and Opportunities. NEUROEPIGENETICS 2015; 1:2-13. [PMID: 25722961 PMCID: PMC4337407 DOI: 10.1016/j.nepig.2014.10.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Long-lived post-mitotic cells, such as the majority of human neurons, must respond effectively to ongoing changes in neuronal stimulation or microenvironmental cues through transcriptional and epigenomic regulation of gene expression. The role of epigenomic regulation in neuronal function is of fundamental interest to the neuroscience community, as these types of studies have transformed our understanding of gene regulation in post-mitotic cells. This perspective article highlights many of the resources available to researchers interested in neuroepigenomic investigations and discusses some of the current obstacles and opportunities in neuroepigenomics.
Collapse
Affiliation(s)
- John S. Satterlee
- National Institute on Drug Abuse (NIDA), Division of Basic Neurobiology and Behavioral Research, 6001 Executive Boulevard, Bethesda, MD 20850, USA
| | - Andrea Beckel-Mitchener
- National Institute on Mental Health (NIMH), Division of Neuroscience and Basic Behavioral Science, 6001 Executive Boulevard Bethesda, MD 20892-9641, USA
| | - Roger Little
- National Institute on Drug Abuse (NIDA), Division of Basic Neurobiology and Behavioral Research, 6001 Executive Boulevard, Bethesda, MD 20850, USA
| | - Dena Procaccini
- National Institute on Drug Abuse (NIDA), Division of Basic Neurobiology and Behavioral Research, 6001 Executive Boulevard, Bethesda, MD 20850, USA
| | - Joni L. Rutter
- National Institute on Drug Abuse (NIDA), Division of Basic Neurobiology and Behavioral Research, 6001 Executive Boulevard, Bethesda, MD 20850, USA
| | - Amy C. Lossie
- Office of Behavioral and Social Sciences Research (OBSSR), Division of Program Coordination, Planning, and Strategic Initiatives, Office of the Director/National Institutes of Health (NIH), 31 Center Drive, Bethesda, MD 20892, USA
| |
Collapse
|
8
|
Li F, Wang Y, Zhang G, Zhou J, Yang L, Guan H. Expression and methylation of DNA repair genes in lens epithelium cells of age-related cataract. Mutat Res 2014; 766-767:31-6. [PMID: 25847269 DOI: 10.1016/j.mrfmmm.2014.05.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2014] [Revised: 05/21/2014] [Accepted: 05/30/2014] [Indexed: 02/05/2023]
Abstract
The development of age-related cataract (ARC) is associated with DNA damage of the lens epithelial cells (LECs). This study aimed to investigate the expression level of DNA repair genes in LECs of ARC and examine whether any altered expression observed could result from DNA methylation of the promoter region of the genes. The expression levels of DNA repair genes were evaluated by microarray analysis. The results were further confirmed by qRT-PCR. DNA methylation of genes with altered expression was determined by bisulfite-specific (BSP) PCR. The mRNA levels of 10 DNA repair genes were decreased and the level of 1 DNA repair gene was increased in LECs of ARC patients compared with controls. The promoter region of the MGMT gene was hypermethylated in ARC tissue compared to controls. The data provide evidence that altered expression of DNA repair genes is associated with pathogenesis of ARC. DNA methylation of MGMT may regulate the expression of the gene and be involved in the development of ARC.
Collapse
Affiliation(s)
- Fei Li
- Eye Institute, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Yong Wang
- Eye Institute, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Guowei Zhang
- Eye Institute, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Jing Zhou
- Eye Institute, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Ling Yang
- Eye Institute, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Huaijin Guan
- Eye Institute, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China.
| |
Collapse
|