1
|
Ye HF, Zhang X, Zhao ZN, Zheng C, Fei P, Xu Y, Lyu J, Chen JL, Guo XX, Zhu H, Zhao PQ. Characterization of N 6-methyladenosine long non-coding RNAs in sporadic congenital cataract and age-related cataract. Int J Ophthalmol 2024; 17:1973-1986. [PMID: 39559306 PMCID: PMC11528264 DOI: 10.18240/ijo.2024.11.02] [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: 11/15/2023] [Accepted: 07/09/2024] [Indexed: 11/20/2024] Open
Abstract
AIM To characterize the N6-methyladenosine (m6A) modification patterns in long non-coding RNAs (lncRNAs) in sporadic congenital cataract (CC) and age-related cataract (ARC). METHODS Anterior capsule of the lens were collected from patients with CC and ARC. Methylated RNA immunoprecipitation with next-generation sequencing and RNA sequencing were performed to identify m6A-tagged lncRNAs and lncRNAs expression. Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyses and Gene Ontology annotation were used to predict potential functions of the m6A-lncRNAs. RESULTS Large amount of m6A peaks within lncRNA were identified for both CC and ARC, while the level was much higher in ARC (49 870 peaks) than that in CC (18 688 peaks), yet those difference between ARC in younger age group (ARC-1) and ARC in elder age group (ARC-2) was quite slight. A total of 1305 hypermethylated and 1178 hypomethylated lncRNAs, as well as 182 differential expressed lncRNAs were exhibited in ARC compared with CC. On the other hand, 5893 hypermethylated and 5213 hypomethylated lncRNAs, as well as 155 significantly altered lncRNA were identified in ARC-2 compared with ARC-1. Altered lncRNAs in ARC were mainly associated with the organization and biogenesis of intracellular organelles, as well as nucleotide excision repair. CONCLUSION Our results for the first time present an overview of the m6A methylomes of lncRNA in CC and ARC, providing a solid basis and uncovering a new insight to reveal the potential pathogenic mechanism of CC and ARC.
Collapse
Affiliation(s)
- Hong-Fei Ye
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Xiang Zhang
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Zhen-Nan Zhao
- Department of Ophthalmology and Vision Science, Eye Ear Nose and Throat Hospital of Fudan University, Shanghai 200031, China
| | - Ce Zheng
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Ping Fei
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Yu Xu
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Jiao Lyu
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Ji-Li Chen
- Department of Ophthalmology, Shanghai Shibei Hospital of Jing'an District, Shanghai 200040, China
| | - Xun-Xiang Guo
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Huang Zhu
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Pei-Quan Zhao
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| |
Collapse
|
2
|
Wang L, Zhu D, Yang Y, He Y, Sun J, Li YM, Wang ZJ, Li P. DNA hypermethylation of COL4A1 in ultraviolet-B-induced age-related cataract models in vitro and in vivo. Int J Ophthalmol 2024; 17:1791-1799. [PMID: 39430019 PMCID: PMC11422356 DOI: 10.18240/ijo.2024.10.04] [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: 03/04/2024] [Accepted: 08/20/2024] [Indexed: 10/22/2024] Open
Abstract
AIM To explore the DNA methylation of COL4A1 in ultraviolet-B (UVB)-induced age-related cataract (ARC) models in vitro and in vivo. METHODS Human lens epithelium B3 (HLEB3) cells and Sprague Dawley rats were exposure to UVB respectively. The MTT assay was utilized to evaluate cell proliferation. Flow cytometry was employed for analysis of cell apoptosis and cell cycle. COL4A1 expression in HLEB3 cells and anterior lens capsules were assessed using Western blot and reverse transcription-polymerase chain reaction (RT-PCR). The localization of COL4A1 in HLEB3 cells was determined by immunofluorescence. The methylation status of CpG islands located in COL4A1 promoter was verified using bisulfite-sequencing PCR (BSP). DNMTs and TETs mRNA levels was examined by RT-PCR. RESULTS UVB exposure decreased HLEB3 cells proliferation, while increased the apoptosis rate and cells were arrested in G0/G1 phase. COL4A1 expression was markedly inhibited in UVB treated cells compared to the controls. Hypermethylation status was detected in the CpG islands within COL4A1 promoter in HLEB3 cells subjected to UVB exposure. Expressions of DNMTs including DNMT1/2/3 were elevated in UVB treated HLEB3 cells compared to that in the controls, while expressions of TETs including TET1/2/3 showed the opposite trend. Results from the UVB treated rat model further confirmed the decreased expression of COL4A1, hypermethylation status of the CpG islands at promoter of COL4A1 and abnormal expression of DNMT1/2/3 and TET1/2/in UVB exposure group. CONCLUSION DNA hypermethylation of COL4A1 promoter CpG islands is correlated with decreased COL4A1 expression in UVB induced HLEB3 cells and anterior lens capsules of rats.
Collapse
Affiliation(s)
- Li Wang
- Department of Optometry, Xi'an Medical University, Xi'an 710021, Shaanxi Province, China
| | - Dan Zhu
- Department of Optometry, Xi'an Medical University, Xi'an 710021, Shaanxi Province, China
| | - Yang Yang
- Department of Optometry, Xi'an Medical University, Xi'an 710021, Shaanxi Province, China
| | - Yuan He
- Department of Optometry, Xi'an Medical University, Xi'an 710021, Shaanxi Province, China
- Department of Ophthalmology, the Second Affiliated Hospital of Xi'an Medical University, Xi'an 710038, Shaanxi Province, China
| | - Jing Sun
- Department of Optometry, Xi'an Medical University, Xi'an 710021, Shaanxi Province, China
| | - Yi-Ming Li
- Department of Optometry, Xi'an Medical University, Xi'an 710021, Shaanxi Province, China
| | - Zi-Jing Wang
- Department of Optometry, Xi'an Medical University, Xi'an 710021, Shaanxi Province, China
| | - Peng Li
- Department of Ophthalmology, Xijing 986 Hospital, Fourth Military Medical University, Xi'an 710054, Shaanxi Province, China
| |
Collapse
|
3
|
Research on Werner Syndrome: Trends from Past to Present and Future Prospects. Genes (Basel) 2022; 13:genes13101802. [PMID: 36292687 PMCID: PMC9601476 DOI: 10.3390/genes13101802] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/02/2022] [Accepted: 10/04/2022] [Indexed: 11/17/2022] Open
Abstract
A rare and autosomal recessive premature aging disorder, Werner syndrome (WS) is characterized by the early onset of aging-associated diseases, including shortening stature, alopecia, bilateral cataracts, skin ulcers, diabetes, osteoporosis, arteriosclerosis, and chromosomal instability, as well as cancer predisposition. WRN, the gene responsible for WS, encodes DNA helicase with a 3′ to 5′ exonuclease activity, and numerous studies have revealed that WRN helicase is involved in the maintenance of chromosome stability through actions in DNA, e.g., DNA replication, repair, recombination, and epigenetic regulation via interaction with DNA repair factors, telomere-binding proteins, histone modification enzymes, and other DNA metabolic factors. However, although these efforts have elucidated the cellular functions of the helicase in cell lines, they have not been linked to the treatment of the disease. Life expectancy has improved for WS patients over the past three decades, and it is hoped that a fundamental treatment for the disease will be developed. Disease-specific induced pluripotent stem (iPS) cells have been established, and these are expected to be used in drug discovery and regenerative medicine for WS patients. In this article, we review trends in research to date and present some perspectives on WS research with regard to the application of pluripotent stem cells. Furthermore, the elucidation of disease mechanisms and drug discovery utilizing the vast amount of scientific data accumulated to date will be discussed.
Collapse
|
4
|
Jiang S, Jia Y, Gao Z. LncRNA KCNQ1OT1 promotes apoptosis and oxidative stress of human lens epithelial cells through epigenetic regulation of WRN. Curr Eye Res 2022; 47:I-X. [PMID: 35179402 DOI: 10.1080/02713683.2022.2026975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
PURPOSE Long non-coding RNA KCNQ1OT1 is fundamental to age-related cataract (ARC), whereas the underlying mechanism is still unknown. Here, we explored the possible mechanism of KCNQ1OT1 in ARC. METHODS The expression of KCNQ1OT1 in ARC patients and H2O2-treated human lens epithelial cell line SRA01/04 was detected. Gene and protein expression were examined by quantitative real-time PCR and western blot. Cell viability and apoptosis were detected by CCK-8 assay and flow cytometry. The content of reactive oxygen species (ROS) was assessed by fluorescent probe DCFH-DA. The relationship among KCNQ1OT1, G9a, H3K9me1/2 and WRN was verified by RNA pull down and Chromatin immunoprecipitation. RESULTS KCNQ1OT1 was up-regulated in the anterior lens capsule tissues of ARC patients and H2O2-treated SRA01/04 cells. KCNQ1OT1 overexpression suppressed cell viability and facilitated apoptosis in H2O2-treated SRA01/04 cells. KCNQ1OT1 up-regulation enhanced the levels of ROS and malondialdehyde (MDA), and reduced the levels of superoxide dismutase (SOD) and catalase (CAT) in H2O2-treated SRA01/04 cells. WRN up-regulation led to a result opposite to KCNQ1OT1 overexpression. The influence of WRN up-regulation on cell viability, apoptosis and oxidative stress of SRA01/04 cells was rescued by KCNQ1OT1 overexpression. Additionally, KCNQ1OT1 interacted with G9a. Both G9a and H3K9me1/2 interacted with WRN promoter. G9a deficiency significantly enhanced WRN expression and repressed H3K9me1/2 expression in SRA01/04 cells, which was abrogated by KCNQ1OT1 up-regulation. CONCLUSION This study demonstrated that KCNQ1OT1 promoted apoptosis and oxidative stress of human LECs through G9a-driven epigenetic regulation of WRN. This work highlights a novel lncRNA involving key regulators of ARC.
Collapse
Affiliation(s)
- Shengqun Jiang
- Department of Ophthalmology, The First Affiliated Hospital of Bengbu Medical College, No.287 Changhuai Road, Bengbu 233004, Anhui Province, China
| | - Yanwen Jia
- Eye Institute, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, No 29 Xianglong Lane, Changzhou 211166, Jiangsu Province, China
| | - Ziqing Gao
- Department of Ophthalmology, The First Affiliated Hospital of Bengbu Medical College, No.287 Changhuai Road, Bengbu 233004, Anhui Province, China
| |
Collapse
|
5
|
Gudmundsrud R, Skjånes TH, Gilmour BC, Caponio D, Lautrup S, Fang EF. Crosstalk among DNA Damage, Mitochondrial Dysfunction, Impaired Mitophagy, Stem Cell Attrition, and Senescence in the Accelerated Ageing Disorder Werner Syndrome. Cytogenet Genome Res 2021; 161:297-304. [PMID: 34433164 DOI: 10.1159/000516386] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 12/10/2020] [Indexed: 12/13/2022] Open
Abstract
Werner syndrome (WS) is an accelerated ageing disease caused by multiple mutations in the gene encoding the Werner DNA helicase (WRN). The major clinical features of WS include wrinkles, grey hair, osteoporosis, and metabolic phenomena such as atherosclerosis, diabetes, and fatty liver, and resemble those seen in normal ageing, but occur earlier, in middle age. Defective DNA repair resulting from mutations in WRN explain the majority of the clinical features of WS, but the underlying mechanisms driving the larger metabolic dysfunction remain elusive. Recent studies in animal models of WS and in WS patient cells and blood samples suggest the involvement of impaired mitophagy, NAD+ depletion, and accumulation of damaged mitochondria in metabolic dysfunction. This mini-review summarizes recent progress in the understanding of the molecular mechanisms of metabolic dysfunction in WS, with the involvement of DNA damage, mitochondrial dysfunction, mitophagy reduction, stem cell impairment, and senescence. Future studies on NAD+ and mitophagy may shed light on potential therapeutic strategies for the WS patients.
Collapse
Affiliation(s)
- Ruben Gudmundsrud
- Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital, Lørenskog, Norway
| | - Tarjei H Skjånes
- Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital, Lørenskog, Norway
| | - Brian C Gilmour
- The Norwegian Centre on Healthy Ageing (NO-Age), Oslo, Norway
| | - Domenica Caponio
- Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital, Lørenskog, Norway
| | - Sofie Lautrup
- Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital, Lørenskog, Norway
| | - Evandro F Fang
- Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital, Lørenskog, Norway.,The Norwegian Centre on Healthy Ageing (NO-Age), Oslo, Norway
| |
Collapse
|
6
|
Ahani-Nahayati M, Niazi V, Moradi A, Pourjabbar B, Roozafzoon R, Baradaran-Rafii A, Keshel SH. Cell-based therapy for ocular disorders: A promising frontier. Curr Stem Cell Res Ther 2021; 17:147-165. [PMID: 34161213 DOI: 10.2174/1574888x16666210622124555] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 02/06/2021] [Accepted: 04/19/2021] [Indexed: 11/22/2022]
Abstract
As the ocular disorders causing long-term blindness or optical abnormalities of the ocular tissue affect the quality of life of patients to a large extent, awareness of their corresponding pathogenesis and the earlier detection and treatment need more consideration. Though current therapeutics result in desirable outcomes, they do not offer an inclusive solution for development of visual impairment to blindness. Accordingly, stem cells, because of their particular competencies, have gained extensive attention for application in regenerative medicine of ocular diseases. In the last decades, a wide spectrum of stem cells surrounding mesenchymal stem/stromal cells (MSC), neural stem cells (NSCs), and embryonic/induced pluripotent stem cells (ESCs/iPSCs) accompanied by Müller glia, ciliary epithelia-derived stem cells, and retinal pigment epithelial (RPE) stem cells have been widely investigated to report their safety and efficacy in preclinical models and also human subjects. In this regard, in the first interventions, RPE cell suspensions were successfully utilized to ameliorate visual defects of the patients suffering from age-related macular degeneration (AMD) after subretinal transplantation. Herein, we will explain the pathogenesis of ocular diseases and highlight the novel discoveries and recent findings in the context of stem cell-based therapies in these disorders, focusing on the in vivo reports published during the last decade.
Collapse
Affiliation(s)
- Milad Ahani-Nahayati
- Department of Tissue Engineering and Applied Cell Science, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Vahid Niazi
- Department of Tissue Engineering and Applied Cell Science, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Alireza Moradi
- Department of Physiology, School of Medicine, Iran University of Medical Science, Tehran, Iran
| | - Bahareh Pourjabbar
- Department of Tissue Engineering and Applied Cell Science, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Reza Roozafzoon
- Department of Tissue Engineering and Applied Cell Science, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Science, Tehran, Iran
| | | | - Saeed Heidari Keshel
- Department of Tissue Engineering and Applied Cell Science, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Science, Tehran, Iran
| |
Collapse
|
7
|
Chen X, Li P, Zhang G, Kang L, Qin B, Mao X, Qin M, Cao Y, Wang Y, Guan H. Comprehensive Profiling of Proteome and Ubiquitome Changes in Human Lens Epithelial Cell Line after Ultraviolet-B Irradiation. ACS OMEGA 2020; 5:32171-32182. [PMID: 33376855 PMCID: PMC7758888 DOI: 10.1021/acsomega.0c03088] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 11/20/2020] [Indexed: 05/08/2023]
Abstract
Ultraviolet-B (UVB) is a recognized risk factor for age-related cataract (ARC) and can cause various changes, including ubiquitination, in lens epithelial cells (LECs). However, the relationship between ubiquitination and ARC is unclear. Herein, we used UVB-irradiated human lens epithelial cell line (SRA01/04) representing the cell model of ARC to investigate the profile changes in the proteome and ubiquitome. A total of 552 differentially expressed proteins (DEPs) and 871 differentially ubiquitinated proteins (DUPs) were identified, and 9 ubiquitination motifs were found. Bioinformatics analysis revealed diverse pathways and biological processes of differential proteins and several DNA damage repair proteins that were potentially mediated via ubiquitin-proteasome pathway. We validated the decreased protein expression of DNA-directed RNA polymerase II subunit RPB2 (POLR2B) in both human cataract capsule tissues and UVB-treated SRA01/04 cells and found that treatment with proteasome inhibitor (MG-132) could reverse the protein level of POLR2B in UVB-irradiated SRA01/04 cells. Our data provide novel information regarding protein expressions and ubiquitination modifications in UVB-induced oxidative damage model. This study might offer a cell-level reference to further investigate the pathogenesis of ARC.
Collapse
|
8
|
Jiang S, Chen J. WRN inhibits oxidative stress-induced apoptosis of human lensepithelial cells through ATM/p53 signaling pathway and its expression is downregulated by DNA methylation. Mol Med 2020; 26:68. [PMID: 32640976 PMCID: PMC7341633 DOI: 10.1186/s10020-020-00187-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 06/16/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Apoptosis and oxidative stress are the main etiology of age related cataract (ARC). This article aims to investigate the role of WRN in lens epithelial cells (LECs). METHODS We estimated the methylation level of WRN in anterior lens capsule tissues of ARC patients. SRA01/04 (LECs) cells were treated with H2O2 or combined with 5-aza-2-deoxycytidine (5-Aza-CdR) or chloroquine. CCK8 and flow cytometry were performed to explore proliferation and apoptosis. The content of ROS was detected by fluorescent probe DCFH-DA. The gene and protein expression was assessed by quantitative real-time PCR or western blot. RESULTS WRN was down-regulated and the methylation level of WRN was increased in the anterior lens capsule tissues. WRN overexpression and 5-Aza-CdR enhanced proliferation and repressed apoptosis and oxidative stress of SRA01/04 cells. 5-Aza-CdR enhanced WRN expression. WRN knockdown inhibited proliferation and promoted apoptosis and oxidative stress of SRA01/04 cells, which was rescued by 5-Aza-CdR. WRN overexpression and 5-Aza-CdR repressed ATM/p53 signaling pathway. Furthermore, chloroquine inhibited proliferation and promoted apoptosis and oxidative stress of SRA01/04 cells by activating ATM/p53 signaling pathway. The influence conferred by chloroquine was abolished by WRN overexpression. CONCLUSION Our study reveals that DNA methylation mediated WRN inhibits apoptosis and oxidative stress of human LECs through ATM/p53 signaling pathway.
Collapse
Affiliation(s)
- Shengqun Jiang
- Ophthalmology Department, The First Affiliated Hospital of Jinan University Guangzhou, No.601 Huangpu Avenue West, Guangzhou, Guangdong Province, China
- Ophthalmology Department, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui Province, China
| | - Jiansu Chen
- Ophthalmology Department, The First Affiliated Hospital of Jinan University Guangzhou, No.601 Huangpu Avenue West, Guangzhou, Guangdong Province, China.
| |
Collapse
|
9
|
Abstract
Aging, the universal phenomenon, affects human health and is the primary risk factor for major disease pathologies. Progeroid diseases, which mimic aging at an accelerated rate, have provided cues in understanding the hallmarks of aging. Mutations in DNA repair genes as well as in telomerase subunits are known to cause progeroid syndromes. Werner syndrome (WS), which is characterized by accelerated aging, is an autosomal-recessive genetic disorder. Hallmarks that define the aging process include genomic instability, telomere attrition, epigenetic alterations, loss of proteostasis, deregulation of nutrient sensing, mitochondrial dysfunction, cellular senescence, stem cell exhaustion, and altered intercellular communication. WS recapitulates these hallmarks of aging and shows increased incidence and early onset of specific cancers. Genome integrity and stability ensure the normal functioning of the cell and are mainly guarded by the DNA repair machinery and telomeres. WRN, being a RecQ helicase, protects genome stability by regulating DNA repair pathways and telomeres. Recent advances in WS research have elucidated WRN’s role in DNA repair pathway choice regulation, telomere maintenance, resolution of complex DNA structures, epigenetic regulation, and stem cell maintenance.
Collapse
Affiliation(s)
- Raghavendra A Shamanna
- Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Deborah L Croteau
- Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Jong-Hyuk Lee
- Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Vilhelm A Bohr
- Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| |
Collapse
|
10
|
Guastafierro T, Bacalini MG, Marcoccia A, Gentilini D, Pisoni S, Di Blasio AM, Corsi A, Franceschi C, Raimondo D, Spanò A, Garagnani P, Bondanini F. Genome-wide DNA methylation analysis in blood cells from patients with Werner syndrome. Clin Epigenetics 2017; 9:92. [PMID: 28861129 PMCID: PMC5577832 DOI: 10.1186/s13148-017-0389-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 08/15/2017] [Indexed: 12/15/2022] Open
Abstract
Background Werner syndrome is a progeroid disorder characterized by premature age-related phenotypes. Although it is well established that autosomal recessive mutations in the WRN gene is responsible for Werner syndrome, the molecular alterations that lead to disease phenotype remain still unidentified. Results To address whether epigenetic changes can be associated with Werner syndrome phenotype, we analysed genome-wide DNA methylation profile using the Infinium MethylationEPIC BeadChip in the whole blood from three patients affected by Werner syndrome compared with three age- and sex-matched healthy controls. Hypermethylated probes were enriched in glycosphingolipid biosynthesis, FoxO signalling and insulin signalling pathways, while hypomethylated probes were enriched in PI3K-Akt signalling and focal adhesion pathways. Twenty-two out of 47 of the differentially methylated genes belonging to the enriched pathways resulted differentially expressed in a publicly available dataset on Werner syndrome fibroblasts. Interestingly, differentially methylated regions identified CERS1 and CERS3, two members of the ceramide synthase family. Moreover, we found differentially methylated probes within ITGA9 and ADAM12 genes, whose methylation is altered in systemic sclerosis, and within the PRDM8 gene, whose methylation is affected in dyskeratosis congenita and Down syndrome. Conclusions DNA methylation changes in the peripheral blood from Werner syndrome patients provide new insight in the pathogenesis of the disease, highlighting in some cases a functional correlation of gene expression and methylation status. Electronic supplementary material The online version of this article (doi:10.1186/s13148-017-0389-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- T Guastafierro
- UOC of Clinical Biochemistry, Sandro Pertini Hospital, Rome, Italy.,CRIIS (Interdisciplinary, Interdepartmental and Specialistic Reference Center for Early Diagnosis of Scleroderma, Treatment of Sclerodermic Ulcers and Videocapillaroscopy), Sandro Pertini Hospital, Rome, Italy
| | - M G Bacalini
- IRCCS Institute of Neurological Sciences, Bologna, Italy
| | - A Marcoccia
- CRIIS (Interdisciplinary, Interdepartmental and Specialistic Reference Center for Early Diagnosis of Scleroderma, Treatment of Sclerodermic Ulcers and Videocapillaroscopy), Sandro Pertini Hospital, Rome, Italy.,UOSD Ischemic Microangiopathy and Sclerodermic Ulcers, Sandro Pertini Hospital, Rome, Italy
| | - D Gentilini
- Centre for Biomedical Research and Technologies, Italian Auxologic Institute, IRCCS, Milan, Italy
| | - S Pisoni
- Centre for Biomedical Research and Technologies, Italian Auxologic Institute, IRCCS, Milan, Italy
| | - A M Di Blasio
- Centre for Biomedical Research and Technologies, Italian Auxologic Institute, IRCCS, Milan, Italy
| | - A Corsi
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - C Franceschi
- IRCCS Institute of Neurological Sciences, Bologna, Italy.,Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy.,Interdepartmental Center "L. Galvani", University of Bologna, Bologna, Italy
| | - D Raimondo
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - A Spanò
- UOC of Clinical Biochemistry, Sandro Pertini Hospital, Rome, Italy
| | - P Garagnani
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy.,Interdepartmental Center "L. Galvani", University of Bologna, Bologna, Italy.,Center for Applied Biomedical Research (CRBA), St. Orsola-Malpighi University Hospital, Bologna, Italy.,Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institute at Huddinge University Hospital, S-141 86 Stockholm, Sweden.,CNR Institute for Molecular Genetics, Unit of Bologna, Bologna, Italy.,Laboratory of Musculoskeletal Cell Biology, Rizzoli Orthopedic Institute, Bologna, Italy
| | - F Bondanini
- CRIIS (Interdisciplinary, Interdepartmental and Specialistic Reference Center for Early Diagnosis of Scleroderma, Treatment of Sclerodermic Ulcers and Videocapillaroscopy), Sandro Pertini Hospital, Rome, Italy.,UOC of Clinical Pathology, Saint' Eugenio Hospital, Rome, Italy
| |
Collapse
|
11
|
Wang Y, Guan H. The Role of DNA Methylation in Lens Development and Cataract Formation. Cell Mol Neurobiol 2017; 37:979-984. [PMID: 27858287 PMCID: PMC11482102 DOI: 10.1007/s10571-016-0447-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Accepted: 11/10/2016] [Indexed: 10/20/2022]
Abstract
Epigenetics pertains to heritable alterations in genomic structural modifications without altering genomic DNA sequence. The studies of epigenetic mechanisms include DNA methylation, histone modifications, and microRNAs. DNA methylation may contribute to silencing gene expression which is a major mechanism of epigenetic gene regulation. DNA methylation regulatory mechanisms in lens development and pathogenesis of cataract represent exciting areas of research that have opened new avenues for association with aging and environment. This review addresses our current understanding of the major mechanisms and function of DNA methylation in lens development, age-related cataract, secondary cataract, and complicated cataract. By understanding the role of DNA methylation in the lens disease and development, it is expected to open up a new therapeutic approach to clinical treatment of cataract.
Collapse
Affiliation(s)
- Yong Wang
- Eye Institute, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, Jiangsu, China
| | - Huaijin Guan
- Eye Institute, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, Jiangsu, China.
| |
Collapse
|
12
|
Cheung HH, Pei D, Chan WY. Stem cell aging in adult progeria. ACTA ACUST UNITED AC 2015; 4:6. [PMID: 26435834 PMCID: PMC4592574 DOI: 10.1186/s13619-015-0021-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 08/29/2015] [Indexed: 12/21/2022]
Abstract
Aging is considered an irreversible biological process and also a major risk factor for a spectrum of geriatric diseases. Advanced age-related decline in physiological functions, such as neurodegeneration, development of cardiovascular disease, endocrine and metabolic dysfunction, and neoplastic transformation, has become the focus in aging research. Natural aging is not regarded as a programmed process. However, accelerated aging due to inherited genetic defects in patients of progeria is programmed and resembles many aspects of natural aging. Among several premature aging syndromes, Werner syndrome (WS) and Hutchinson–Gilford progeria syndrome (HGPS) are two broadly investigated diseases. In this review, we discuss how stem cell aging in WS helps us understand the biology of aging. We also discuss briefly how the altered epigenetic landscape in aged cells can be reversed to a “juvenile” state. Lastly, we explore the potential application of the latest genomic editing technique for stem cell-based therapy and regenerative medicine in the context of aging.
Collapse
Affiliation(s)
- Hoi-Hung Cheung
- CUHK-CAS GIBH Joint Research Laboratory on Stem Cell and Regenerative Medicine, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong S.A.R., China
| | - Duanqing Pei
- Chinese Academy of Sciences (CAS) Guangzhou Institutes of Biomedicine and Health (GIBH), Guangzhou, China
| | - Wai-Yee Chan
- CUHK-CAS GIBH Joint Research Laboratory on Stem Cell and Regenerative Medicine, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong S.A.R., China ; The Chinese University of Hong Kong, Room G03A, Lo Kwee-Seong Intergrated Biomedical Science Building, Shatin, N.T., Hong Kong S.A.R., China
| |
Collapse
|