1
|
Ding R, Chen C, Wang L, Wang Y, Chai Z, He S, Zhang Q, Cheng S, Zou R. Matrix Stiffness Regulates the Osteogenic Differentiation of hPDLSCs via DNA Methylation. Int Dent J 2025; 75:100783. [PMID: 40315698 PMCID: PMC12090244 DOI: 10.1016/j.identj.2025.02.022] [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: 01/05/2025] [Revised: 02/13/2025] [Accepted: 02/24/2025] [Indexed: 05/04/2025] Open
Abstract
OBJECTIVES This study aimed to examine the influence of matrix stiffness on osteogenic differentiation via epigenetic mechanisms in human periodontal ligament stem cells (hPDLSCs), with implications for understanding orthodontic tooth movement. MATERIALS AND METHODS hPDLSCs were cultured on substrates with varying stiffness (soft and stiff). Dot blot and immunofluorescence techniques were employed to measure global DNA methylation levels. RT-qPCR and alkaline phosphatase (ALP) activity assays were conducted to assess differences in DNA methylation and osteogenic potential. Additionally, ELISA was used to quantify DNA methyltransferase content and activity. RESULTS hPDLSCs on stiffer substrates exhibited increased 5-methylcytosine (5-mC) and higher global DNA methylation levels than those on soft substrates. With increased matrix stiffness, DNMT3A and DNMT3B mRNA expression levels rose. hPDLSCs on stiff matrices also showed elevated DNMT3B enzyme content and osteogenic activity. When global DNA methylation was reduced, mRNA levels of RUNX2, ALP, and Col-1 decreased, along with a notable reduction in ALP staining intensity in the inhibitor group. CONCLUSIONS Matrix stiffness is positively associated with global DNA methylation, with DNMT3B likely mediating this regulation in hPDLSCs. Furthermore, DNA methylation levels are positively linked to the osteogenic capability of hPDLSCs.
Collapse
Affiliation(s)
- Rong Ding
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China; Clinical Research Centre of Shaanxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China; College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Chen Chen
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China; Clinical Research Centre of Shaanxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China; College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Ling Wang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China; Clinical Research Centre of Shaanxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China; College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Yijie Wang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China; Clinical Research Centre of Shaanxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China; College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Zhen Chai
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China; Clinical Research Centre of Shaanxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China; College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Siyu He
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China; Clinical Research Centre of Shaanxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China; College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Qianqian Zhang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China; Clinical Research Centre of Shaanxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China; College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Shuli Cheng
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China; Clinical Research Centre of Shaanxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China; College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China.
| | - Rui Zou
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China; Clinical Research Centre of Shaanxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China; College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China.
| |
Collapse
|
2
|
KONTIC MILICA, MARKOVIC FILIP. Use of DNA methylation patterns for early detection and management of lung cancer: Are we there yet? Oncol Res 2025; 33:781-793. [PMID: 40191732 PMCID: PMC11964873 DOI: 10.32604/or.2024.057231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 11/20/2024] [Indexed: 04/09/2025] Open
Abstract
Detecting lung cancer early is crucial for improving survival rates, yet it remains a significant challenge due to many cases being diagnosed at advanced stages. This review aims to provide advances in epigenetics which have highlighted DNA methylation patterns as promising biomarkers for early detection, prognosis, and treatment response in lung cancer. Techniques like bisulfite conversion followed by PCR, digital droplet polymerase chain reaction, and next-generation sequencing are commonly used for detecting these methylation patterns, which occur early in the cancer development process and can be detected in non-invasive samples like blood and sputum. Key genes such as SHOX2 and RASSF1A have demonstrated high sensitivity and specificity in clinical studies, making them crucial for diagnostic purposes. However, several challenges remain to be overcome before these biomarkers can be widely adopted for use in clinical practice. Standardizing the assays and validating their effectiveness are critical steps. Additionally, integrating methylation biomarkers with existing diagnostic tools could significantly enhance the accuracy of lung cancer detection, providing a more comprehensive diagnostic approach. Although progress has been made in understanding and utilizing DNA methylation patterns for lung cancer detection, more research and extensive clinical trials are necessary to fully harness their potential. These efforts will help establish the robustness of methylation patterns as biomarkers and therapeutic targets, ultimately leading to better prevention, diagnosis, and treatment strategies for lung cancer. In conclusion, DNA methylation states represent a promising avenue for advancing early detection, accurate diagnosis, and management of lung cancer.
Collapse
Affiliation(s)
- MILICA KONTIC
- Clinic for Pulmonology, University Clinical Center of Serbia, Belgrade, 11000, Serbia
- School of Medicine, University of Belgrade, Belgrade, 11000, Serbia
| | - FILIP MARKOVIC
- Clinic for Pulmonology, University Clinical Center of Serbia, Belgrade, 11000, Serbia
| |
Collapse
|
3
|
Lee R, Ong J, Waisberg E, Mader T, Berdahl J, Suh A, Panzo N, Memon H, Sampige R, Katsev B, Kadipasaoglu CM, Mason CE, Beheshti A, Zwart SR, Smith SM, Lee AG. Potential Risks of Ocular Molecular and Cellular Changes in Spaceflight. Semin Ophthalmol 2025:1-11. [PMID: 40094398 DOI: 10.1080/08820538.2025.2471443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/12/2025] [Accepted: 02/17/2025] [Indexed: 03/19/2025]
Abstract
PURPOSE Many fundamental cellular and molecular changes are known to occur in biological systems during spaceflight, including oxidative stress, DNA damage, mitochondrial damage, epigenetic factors, telomere lengthening, and microbial shifts. We can apply the consequences of these molecular changes in ocular cells, such as the retinal ganglion cells and corneal epithelium, to identify ophthalmologic risks during spaceflight. This review aims to discuss the potential molecular changes in greater detail and apply the principles to ocular cells and ophthalmic disease risk in astronauts. METHODS A targeted, relevant search of the literature on the topic and related topics of ocular surface and spaceflight was conducted with scholarly databases PubMed, Web of Science, and Embase from inception to July2024 with search terms "oxidative stress"; "DNA damage"; "Mitochondrial Dysfunction"; "Epigenetics"; "Telomeres"; "Microbiome"; "ocular cells"; "spaceflight"; "microgravity"; "radiation." RESULTS A total of 115 articles were included following screening and eligibility assessment. Key findings include molecular changes and their contributions to ophthalmic diseases like cataracts, spaceflight-associated neuro-ocular syndrome, and dry eye syndrome. CONCLUSION This review provides a comprehensive overview of risks to vision associated with long-duration spaceflight missions beyond low Earth orbit (LEO). Further investigation into targeted countermeasures is imperative to mitigate vision-threatening sequelae in astronauts undertaking deep-space exploration.
Collapse
Affiliation(s)
- Ryung Lee
- Touro College of Osteopathic Medicine, New York, NY, USA
| | - Joshua Ong
- Department of Ophthalmology and Visual Sciences, University of Michigan Kellogg Eye Center, Ann Arbor, Michigan, USA
| | - Ethan Waisberg
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | | | - John Berdahl
- Vance Thompson Vision, Sioux Falls, South Dakota, USA
| | - Alex Suh
- Tulane School of Medicine, New Orleans, Louisiana, USA
| | | | - Hamza Memon
- Texas A&M School of Medicine, Bryan, TX, USA
| | - Ritu Sampige
- Center for Space Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Blake Katsev
- Department of Ophthalmology and Visual Sciences, Washington University, Saint Louis, MO, USA
| | | | - Christopher E Mason
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY, USA
| | - Afshin Beheshti
- Center for Space Biomedicine, McGowan Institute for Regenerative Medicine, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Sara R Zwart
- Human Health and Performance Directorate, NASA Johnson Space Center, Houston, TX, USA
| | - Scott M Smith
- Human Health and Performance Directorate, NASA Johnson Space Center, Houston, TX, USA
| | - Andrew G Lee
- Department of Ophthalmology, Blanton Eye Institute, Houston Methodist Hospital, Houston, TX, USA
- Departments of Ophthalmology, Neurology, and Neurosurgery, Weill Cornell Medicine, New York, New York, USA
- Department of Ophthalmology, University of Texas Medical Branch, Galveston, TX, USA
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Ophthalmology, The University of Iowa Hospitals and Clinics, Iowa City, Iowa, USA
| |
Collapse
|
4
|
Kang L, Bao S, Li P, Zhang G, Zhu X, Ji M, Guan H. METTL14-mediated depression of NEIL1 aggravates oxidative damage and mitochondrial dysfunction of lens epithelial cells through regulating KEAP1/NRF2 pathways. Cell Signal 2025; 127:111623. [PMID: 39855533 DOI: 10.1016/j.cellsig.2025.111623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 01/27/2025]
Abstract
Abnormal base excision repair (BER) pathway and N6-methyladenosine (m6A) of RNA have been proved to be significantly related to age-related cataract (ARC) pathogenesis. However, the relationship between the Nei Endonuclease VIII-Like1 (NEIL1) gene (a representative DNA glycosylase of BER pathway) and its m6A modification remains unclear. Here, we showed that the expression of NEIL1 was decreased in the ARC anterior lens capsules and H2O2-stimulated SRA01/04 cells. Our findings demonstrated that ectopic expression of NEIL1 alleviated DNA oxidative damage, apoptosis and mitochondrial dysfunction through disturbing KEAP1/NRF2 interaction. Furthermore, silencing NEIL1 aggravated H2O2-induced lens opacity, whereas ML334 could mitigate lens cloudy ex vitro in rat lenses. Besides, intravitreal injection of AAV2-NEIL1 alleviated lens opacity in Emory mice in vivo. Mechanistically, the N(6)-Methyladenosine (m6A) methyltransferase-like 14 (METTL14) was identified as a factor in promoting m6A modification of NEIL1, which resulted in the recruitment of YTHDF2 to recognize and impair NEIL1 RNA stability. Collectively, these findings highlight the critical role of the m6A modification in NEIL1 on regulating oxidative stress and mitochondrial homeostasis through KEAP1/NRF2 pathways, providing a new way to explore the pathogenesis of ARC.
Collapse
Affiliation(s)
- Lihua Kang
- Eye Institute, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Sijie Bao
- Eye Institute, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Pengfei Li
- Eye Institute, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Guowei Zhang
- Eye Institute, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Xi Zhu
- Eye Institute, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Min Ji
- Eye Institute, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China.
| | - Huaijin Guan
- Eye Institute, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China.
| |
Collapse
|
5
|
Shetty R, Khamar P, Kannan R, Thacker P, Kumar NR, Ghosh A, Deshpande V. Epigenetic Modulation Directs Recovery Post LASIK and SMILE Surgery: An Experimental Study. Life (Basel) 2025; 15:246. [PMID: 40003656 PMCID: PMC11856829 DOI: 10.3390/life15020246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/22/2025] [Accepted: 01/31/2025] [Indexed: 02/27/2025] Open
Abstract
PURPOSE refractive surgery, such as LASIK and SMILE, induces a wound healing response that leads to significant corneal stromal remodeling. We have shown that the protein profile in the stroma changes dramatically immediately post-surgery. However, the methylation status of the DNA post-refractive surgery remains unknown. DESIGN/PARTICIPANTS DNA methylation study. Refractive surgery (SMILE/LASIK) performed on donor eye globes. METHOD we investigated the epigenetic changes post-surgery in relation to long term ECM remodeling in an experimental ex vivo study design. Donor globes (n = 19) were obtained from the eye bank. Three globes served as non-surgical controls while SMILE (-6DS) and LASIK surgery (-6DS) were performed on eight globes each and incubated for 3 days and 2 weeks (n = 4 per group per time point). Here, we compared the DNA methylation landscapes of LASIK and SMILE stroma using the Illumina Infinium Human Methylation 850 EPIC array (HM850). RESULTS significant changes in DNA methylation patterns were observed post-operatively in both LASIK and SMILE groups. Specific genes involved in the activation of actin cytoskeleton and inflammation (smad3, prkca and ssh2) showed hypomethylation in LASIK after 2 weeks and LASIK SMILE after 3 days, respectively, suggesting their active role in corneal repair. The genes (gaa, gstm1, mgat1, galnt9 and galnt5) involved in sphingolipid metabolism and mucin biosynthesis showed hypomethylation in SMILE after 3 days. CONCLUSIONS our results suggest that altered DNA methylation patterns may have relevance to the development of complications of haze post-refractive surgery. It also presents the opportunity to utilize drugs that regulate chromatin remodeling for optimal outcomes.
Collapse
Affiliation(s)
- Rohit Shetty
- Cornea and Refractive Services, Narayana Nethralaya, Bangalore 560010, India; (R.S.); (P.K.)
| | - Pooja Khamar
- Cornea and Refractive Services, Narayana Nethralaya, Bangalore 560010, India; (R.S.); (P.K.)
| | - Ramaraj Kannan
- GROW Research Laboratory, Narayana Netralaya Foundation, Bangalore 560099, India; (R.K.); (P.T.); (N.R.K.)
| | - Puja Thacker
- GROW Research Laboratory, Narayana Netralaya Foundation, Bangalore 560099, India; (R.K.); (P.T.); (N.R.K.)
| | - Nimisha Rajiv Kumar
- GROW Research Laboratory, Narayana Netralaya Foundation, Bangalore 560099, India; (R.K.); (P.T.); (N.R.K.)
| | - Arkasubhra Ghosh
- GROW Research Laboratory, Narayana Netralaya Foundation, Bangalore 560099, India; (R.K.); (P.T.); (N.R.K.)
| | - Vrushali Deshpande
- GROW Research Laboratory, Narayana Netralaya Foundation, Bangalore 560099, India; (R.K.); (P.T.); (N.R.K.)
| |
Collapse
|
6
|
Hu Y, Su D, Zhang Y, Fu Y, Li S, Chen X, Zhang X, Zheng S, Ma X, Hu S. Genome-wide DNA methylation and transcriptome sequencing analyses of lens tissue in an age-related mouse cataract model. PLoS One 2025; 20:e0316766. [PMID: 39883715 PMCID: PMC11781636 DOI: 10.1371/journal.pone.0316766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 12/15/2024] [Indexed: 02/01/2025] Open
Abstract
DNA methylation is known to be associated with cataracts. In this study, we used a mouse model and performed DNA methylation and transcriptome sequencing analyses to find epigenetic indicators for age-related cataracts (ARC). Anterior lens capsule membrane tissues from young and aged mice were analyzed by MethylRAD-seq to detect the genome-wide methylation of extracted DNA. The young and aged mice had 76,524 and 15,608 differentially methylated CCGG and CCWGG sites, respectively. The Pearson correlation analysis detected 109 and 33 differentially expressed genes (DEGs) with negative methylation at CCGG and CCWGG sites, respectively, in their promoter regions. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) functional enrichment analyses showed that DEGs with abnormal methylation at CCGG sites were primarily associated with protein kinase C signaling (Akap12, Capzb), protein threonine kinase activity (Dmpk, Mapkapk3), and calcium signaling pathway (Slc25a4, Cacna1f), whereas DEGs with abnormal methylation at CCWGG sites were associated with ribosomal protein S6 kinase activity (Rps6ka3). These genes were validated by pyrosequencing methylation analysis. The results showed that the ARC group (aged mice) had lower Dmpk and Slc25a4 methylation levels and a higher Rps6ka3 methylation than the control group (young mice), which is consistent with the results of the joint analysis of differentially methylated and differentially expressed genes. In conclusion, we confirmed the genome-wide DNA methylation pattern and gene expression profile of ARC based on the mouse cataract model with aged mice. The identified methylation molecular markers have great potential for application in the future diagnosis and treatment of ARC.
Collapse
Affiliation(s)
- Yuzhu Hu
- Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Dongmei Su
- Department of Genetics, Health Department, National Research Institute for Family Planning, Beijing, China
- Graduate School, Peking Union Medical College, Beijing, China
| | - Yue Zhang
- Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Yanjiang Fu
- Daqing Ophthalmology Hospital, Daqing, Heilongjiang, China
| | - Sijia Li
- Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Xiaoya Chen
- Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Xiao Zhang
- Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Shunfei Zheng
- Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Xu Ma
- Department of Genetics, Health Department, National Research Institute for Family Planning, Beijing, China
- Graduate School, Peking Union Medical College, Beijing, China
| | - Shanshan Hu
- Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| |
Collapse
|
7
|
Oncu S, Becit-Kizilkaya M, Bilir A, Saritas A, Arikan-Soylemez ES, Koca HB, Firat F, Ugur-Kaplan AB, Yilmaz MA. Anti-Cataract Effect of the Traditional Aqueous Extract of Yerba Mate ( Ilex paraguariensis A. St.-Hil.): An In Ovo Perspective. Life (Basel) 2024; 14:994. [PMID: 39202736 PMCID: PMC11355064 DOI: 10.3390/life14080994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 08/07/2024] [Accepted: 08/08/2024] [Indexed: 09/03/2024] Open
Abstract
INTRODUCTION The therapeutic effect of different doses of the traditional aqueous extract of dried leaves of yerba mate (Ilex paraguariensis A. St.-Hil.) was investigated in an experimental cataract model in chicken embryos. METHODS AND RESULTS LC-MS/MS analysis allowed the identification and quantification of 53 metabolites. In the hydrocortisone-induced cataract model, lenses were examined morphologically after treatment and parameters related to oxidative stress (total antioxidant/oxidant status (TAS/TOS), glutathione (GSH), and malondialdehyde (MDA)) were evaluated. Antiproliferative cell nuclear antigen (PCNA) and caspase-3 H-scores were determined and crystallin alpha A (CRYAA) gene expression in the lenses was measured by RT-PCR. The degree of cataract decreased in all treatment groups. While there was no significant difference in TAS levels compared to the negative control, TOS, GSH, and MDA levels were dose-dependently regulated. Treatment groups other than the high-dose group regulated the decrease in PCNA and the increase in caspase-3. CRYAA gene expression increased significantly only at the lowest dose. CONCLUSION YM, which is becoming increasingly popular as a traditional tea, showed a therapeutic effect on hydrocortisone-induced cataracts in chicken embryos at relatively low doses.
Collapse
Affiliation(s)
- Seyma Oncu
- Department of Medical Pharmacology, Faculty of Medicine, Afyonkarahisar Health Sciences University, Afyonkarahisar 03030, Turkey
| | - Merve Becit-Kizilkaya
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Afyonkarahisar Health Sciences University, Afyonkarahisar 03030, Turkey
| | - Abdulkadir Bilir
- Department of Anatomy, Faculty of Medicine, Afyonkarahisar Health Sciences University, Afyonkarahisar 03030, Turkey
| | - Alperen Saritas
- Department of Anatomy, Faculty of Medicine, Afyonkarahisar Health Sciences University, Afyonkarahisar 03030, Turkey
| | - Evrim Suna Arikan-Soylemez
- Department of Medical Biology, Faculty of Medicine, Afyonkarahisar Health Sciences University, Afyonkarahisar 03030, Turkey
| | - Halit Bugra Koca
- Department of Biochemistry, Faculty of Medicine, Afyonkarahisar Health Sciences University, Afyonkarahisar 03030, Turkey
| | - Fatma Firat
- Department of Histology and Embryology, Medicine Faculty, Afyonkarahisar Health Sciences University, Afyonkarahisar 03030, Turkey
| | - Afife Busra Ugur-Kaplan
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Ataturk University, Erzurum 25240, Turkey
| | - Mustafa Abdullah Yilmaz
- Department of Analytical Chemistry, Faculty of Pharmacy, Dicle University, Diyarbakir 21280, Turkey
| |
Collapse
|
8
|
Chang W, Zhao Y, Rayêe D, Xie Q, Suzuki M, Zheng D, Cvekl A. Dynamic changes in whole genome DNA methylation, chromatin and gene expression during mouse lens differentiation. Epigenetics Chromatin 2023; 16:4. [PMID: 36698218 PMCID: PMC9875507 DOI: 10.1186/s13072-023-00478-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 01/17/2023] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Cellular differentiation is marked by temporally and spatially coordinated gene expression regulated at multiple levels. DNA methylation represents a universal mechanism to control chromatin organization and its accessibility. Cytosine methylation of CpG dinucleotides regulates binding of methylation-sensitive DNA-binding transcription factors within regulatory regions of transcription, including promoters and distal enhancers. Ocular lens differentiation represents an advantageous model system to examine these processes as lens comprises only two cell types, the proliferating lens epithelium and postmitotic lens fiber cells all originating from the epithelium. RESULTS Using whole genome bisulfite sequencing (WGBS) and microdissected lenses, we investigated dynamics of DNA methylation and chromatin changes during mouse lens fiber and epithelium differentiation between embryos (E14.5) and newborns (P0.5). Histone H3.3 variant chromatin landscapes were also generated for both P0.5 lens epithelium and fibers by chromatin immunoprecipitation followed by next generation sequencing (ChIP-seq). Tissue-specific features of DNA methylation patterns are demonstrated via comparative studies with embryonic stem (ES) cells and neural progenitor cells (NPCs) at Nanog, Pou5f1, Sox2, Pax6 and Six3 loci. Comparisons with ATAC-seq and RNA-seq data demonstrate that reduced methylation is associated with increased expression of fiber cell abundant genes, including crystallins, intermediate filament (Bfsp1 and Bfsp2) and gap junction proteins (Gja3 and Gja8), marked by high levels of histone H3.3 within their transcribed regions. Interestingly, Pax6-binding sites exhibited predominantly DNA hypomethylation in lens chromatin. In vitro binding of Pax6 proteins showed Pax6's ability to interact with sites containing one or two methylated CpG dinucleotides. CONCLUSIONS Our study has generated the first data on methylation changes between two different stages of mammalian lens development and linked these data with chromatin accessibility maps, presence of histone H3.3 and gene expression. Reduced DNA methylation correlates with expression of important genes involved in lens morphogenesis and lens fiber cell differentiation.
Collapse
Affiliation(s)
- William Chang
- Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Yilin Zhao
- Genetics, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Danielle Rayêe
- Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Qing Xie
- Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Genetics, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- University of California Santa Cruz, Santa Cruz, CA, 95064, USA
| | - Masako Suzuki
- Genetics, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Deyou Zheng
- Genetics, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Neurology and Neuroscience, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Ales Cvekl
- Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
- Genetics, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
| |
Collapse
|
9
|
DNA Methylation in Atrial Fibrillation and Its Potential Role in Precision Medicine. Methods Mol Biol 2021. [PMID: 32710320 DOI: 10.1007/978-1-0716-0904-0_11] [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: 10/29/2023]
Abstract
Atrial fibrillation (AF), a common arrhythmia, can cause many serious consequences, including stroke and even death. The pathological mechanism of AF is very complicated. Epigenetic mechanisms, especially DNA methylation, contribute to the pathogenesis and maintenance of AF. DNA methylation is an important part of epigenetic and plays a significant role in human physiology and pathology. AF patients possess specific methylation sites (e.g., Pitx2c, RASSF1A, SURs, SERCA2a, and LINC00472), which have potential values of being biomarkers and underlie the diagnosis and prognosis of AF. These methylation sites can also benefit accurate treatment of AF. With deeper understanding into the epigenetic mechanisms of AF, the precision medicine for AF has also developed rapidly. In the future, DNA methylation omics and other research methods will be integrated to explore the epigenetic mechanisms in AF.
Collapse
|
10
|
Rajić J, Dinić S, Uskoković A, Arambašić Jovanović J, Tolić A, Đorđević M, Đorđević M, Poznanović G, Mihailović M, Inic-Kanada A, Barisani-Asenbauer T, Grdović N, Vidaković M. DNA methylation of miR-200 clusters promotes epithelial to mesenchymal transition in human conjunctival epithelial cells. Exp Eye Res 2020; 197:108047. [PMID: 32387379 DOI: 10.1016/j.exer.2020.108047] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 04/17/2020] [Accepted: 04/27/2020] [Indexed: 01/09/2023]
Abstract
Epithelial to mesenchymal transition (EMT) contributes to fibrosis associated pathologies including scarring of different ocular tissues. Recently targeting EMT is seen as an appropriate therapeutic approach for different fibrosis related eye diseases such as macular degeneration or glaucoma surgery related fibrosis. Nevertheless, for ocular surface diseases, target genes specific for particular cell type or condition are still undefined. This study aimed to expose the complex regulatory mechanisms that trigger EMT in human conjunctival epithelial (HCjE) cells. EMT was induced by prolonged treatment with two TGF-β isoforms, TGF-β1 and TGF-β2, and their combination. TGF-β1 showed the strongest potential for initiating EMT in HCjE cells, reflected on morphological changes, cell migration and the levels of mRNA expression of different epithelial (CDH1, OCLN, DSP) and mesenchymal (CDH2, FN1, VIM, SNAI1, ZEB2, TWIST1) marker genes. Co-treatment with the DNA demethylating agent 5-Azacytidine (5-AzaC) was capable of stopping the transition of HCjE cells towards a mesenchymal phenotype, based on morphological features, reduced cell mobility and mRNA and protein expression levels of epithelial and mesenchymal marker genes. An EMT qRT-PCR-based array revealed that EMT induced considerable alterations in gene expression, with downregulation of the majority of epithelial marker genes and upregulation of genes specific for the mesenchymal state. The major effect of 5-AzaC treatment was observed as a suppression of mesenchymal marker genes, suggesting the involvement of upstream negative regulator(s) whose promoter demethylation and subsequent expression will in turn promote EMT switch off. The expression level of miRNAs potentially important for EMT induction was determined using qRT-PCR-based array which pointed at members of miR-200 family as main regulators of EMT process in HCjE cells. 5-AzaC treatment induced increased expression of miR-200a, -200b, -200c and miR-141 towards the control level, indicating important role of DNA methylation in their regulation. The DNA methylation status of both miR-200 family clusters, analyzed with high-resolution melting (HRM) and bisulfite sequencing (Bis-Seq), revealed that TGF-β1-induced EMT was accompanied by increase in promoter CpG methylation of both miR-200 loci, which was reverted after 5-AzaC treatment. In conclusion, our results indicate that DNA demethylation of promoters of miR-200 loci is critically important for stopping and reverting the EMT in human conjunctival epithelial cells, suggesting the potential for the development of novel epigenetic-based therapeutic strategies for treating conjunctival conditions associated with EMT.
Collapse
Affiliation(s)
- Jovana Rajić
- Department of Molecular Biology, Institute for Biological Research "Siniša Stanković", National Institute of Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11060, Belgrade, Serbia
| | - Svetlana Dinić
- Department of Molecular Biology, Institute for Biological Research "Siniša Stanković", National Institute of Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11060, Belgrade, Serbia
| | - Aleksandra Uskoković
- Department of Molecular Biology, Institute for Biological Research "Siniša Stanković", National Institute of Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11060, Belgrade, Serbia
| | - Jelena Arambašić Jovanović
- Department of Molecular Biology, Institute for Biological Research "Siniša Stanković", National Institute of Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11060, Belgrade, Serbia
| | - Anja Tolić
- Department of Molecular Biology, Institute for Biological Research "Siniša Stanković", National Institute of Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11060, Belgrade, Serbia
| | - Marija Đorđević
- Department of Molecular Biology, Institute for Biological Research "Siniša Stanković", National Institute of Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11060, Belgrade, Serbia
| | - Miloš Đorđević
- Department of Molecular Biology, Institute for Biological Research "Siniša Stanković", National Institute of Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11060, Belgrade, Serbia
| | - Goran Poznanović
- Department of Molecular Biology, Institute for Biological Research "Siniša Stanković", National Institute of Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11060, Belgrade, Serbia
| | - Mirjana Mihailović
- Department of Molecular Biology, Institute for Biological Research "Siniša Stanković", National Institute of Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11060, Belgrade, Serbia
| | - Aleksandra Inic-Kanada
- Institute of Specific Prophylaxis and Tropical Medicine, Medical University of Vienna, Kinderspitalgasse 15, 1090, Vienna, Austria
| | - Talin Barisani-Asenbauer
- OCUVAC - Center of Ocular Inflammation and Infection, Laura Bassi Centres of Expertise, Medical University of Vienna, Kinderspitalgasse 15, 1090, Vienna, Austria.
| | - Nevena Grdović
- Department of Molecular Biology, Institute for Biological Research "Siniša Stanković", National Institute of Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11060, Belgrade, Serbia.
| | - Melita Vidaković
- Department of Molecular Biology, Institute for Biological Research "Siniša Stanković", National Institute of Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11060, Belgrade, Serbia
| |
Collapse
|
11
|
Braga DL, Mousovich-Neto F, Tonon-da-Silva G, Salgueiro WG, Mori MA. Epigenetic changes during ageing and their underlying mechanisms. Biogerontology 2020; 21:423-443. [PMID: 32356238 DOI: 10.1007/s10522-020-09874-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 03/20/2020] [Indexed: 12/12/2022]
Abstract
As life expectancy increases worldwide, ageing and age-related diseases arise as a major issue for societies around the globe. Understanding the biological mechanisms underlying the ageing process is thus instrumental for the development of efficient interventions aimed to prevent and treat age-related conditions. Current knowledge in the biogerontology field points to epigenetics as a critical component of the ageing process, not only by serving as a bona-fide marker of biological age but also by controlling and conferring inheritability to cellular and organismal ageing. This is reflected by a myriad of evidences demonstrating the relationship between DNA methylation, histone modifications, chromatin remodeling and small non-coding RNAs and several age-related phenotypes. Given the reversibility of epigenetic alterations, epigenetic reprogramming may also be envisioned as a potential approach to treat age-related disorders. Here we review how different types of epigenetic mechanisms are involved in the ageing process. In addition, we highlight how interventions modulate epigenetics and thus promote health- and lifespan.
Collapse
Affiliation(s)
- Deisi L Braga
- Department of Biochemistry and Tissue Biology, University of Campinas, Rua Monteiro Lobato, 255, Campinas, São Paulo, 13083-862, Brazil
- Program in Genetics and Molecular Biology, University of Campinas, Campinas, São Paulo, 13083-862, Brazil
| | - Felippe Mousovich-Neto
- Department of Biochemistry and Tissue Biology, University of Campinas, Rua Monteiro Lobato, 255, Campinas, São Paulo, 13083-862, Brazil
| | - Guilherme Tonon-da-Silva
- Department of Biochemistry and Tissue Biology, University of Campinas, Rua Monteiro Lobato, 255, Campinas, São Paulo, 13083-862, Brazil
- Program in Genetics and Molecular Biology, University of Campinas, Campinas, São Paulo, 13083-862, Brazil
| | - Willian G Salgueiro
- Department of Biochemistry and Tissue Biology, University of Campinas, Rua Monteiro Lobato, 255, Campinas, São Paulo, 13083-862, Brazil
- Program in Genetics and Molecular Biology, University of Campinas, Campinas, São Paulo, 13083-862, Brazil
| | - Marcelo A Mori
- Department of Biochemistry and Tissue Biology, University of Campinas, Rua Monteiro Lobato, 255, Campinas, São Paulo, 13083-862, Brazil.
- Obesity and Comorbidities Research Center (OCRC), University of Campinas, Campinas, São Paulo, 13083-862, Brazil.
- Experimental Medicine Research Cluster (EMRC), University of Campinas, Campinas, São Paulo, 13083-862, Brazil.
| |
Collapse
|
12
|
Chen E, Bohm K, Rosenblatt M, Kang K. Epigenetic regulation of anterior segment diseases and potential therapeutics. Ocul Surf 2020; 18:383-395. [PMID: 32344150 DOI: 10.1016/j.jtos.2020.04.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 01/31/2020] [Accepted: 04/03/2020] [Indexed: 12/22/2022]
Abstract
In recent years, technological advances in sequencing have accelerated our understanding of epigenetics in ocular development and ophthalmic diseases. We now know that epigenetic modifications are necessary for normal ocular development and biological processes such as corneal wound healing and ocular surface repair, while aberrant epigenetic regulation underlies the pathogenesis of a wide range of ocular diseases, including cataracts and various diseases of the ocular surface. As the epigenetics of the eye is a constantly changing field of medicine, this comprehensive review focuses on innovations and scientific discoveries related to epigenetic control of anterior segment diseases that were published in the English literature in the past five years. These recent studies attempt to elucidate therapeutic targets for the anterior segment pathological processes. Already, recent studies have shown therapeutic potential in targeting epigenetic mechanisms of ocular diseases, and new epigenetic therapies are on the verge of being introduced to clinical practice. New drug targets can potentially emerge as we make further discoveries within this field.
Collapse
Affiliation(s)
- Eric Chen
- Illinois Eye and Ear Infirmary, Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, United States
| | - Kelley Bohm
- Illinois Eye and Ear Infirmary, Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, United States
| | - Mark Rosenblatt
- Illinois Eye and Ear Infirmary, Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, United States
| | - Kai Kang
- Illinois Eye and Ear Infirmary, Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, United States.
| |
Collapse
|
13
|
Krüger-Genge A, Mauger O, Storsberg J, Schmidt C. Epigenetics-Shedding Light on the Path Ahead for Material Sciences. Diseases 2019; 7:diseases7020043. [PMID: 31208004 PMCID: PMC6631476 DOI: 10.3390/diseases7020043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 06/11/2019] [Accepted: 06/13/2019] [Indexed: 11/28/2022] Open
Abstract
The harmonious regulation of bodily function is a necessity for healthy individuals. Looking from the viewpoint of material sciences, one can only marvel at the cellular factories, their renewal, and the overall control of messaging and control of responses. As aging progresses and/or pathologies arise, clinicians may be forced to look for replacement of organs/tissues with medical devices. Since all devices are tailored, a detailed understanding of developmental processes, including aberrant processes leading to pathologies, is crucial to provide clinicians with a suitable device. Although research in the field of epigenetics has produced effective therapeutics and diagnostic markers, our currently fragmented understanding of epigenetic processes as they relate to material development is inherently limited, with logical implications for the success of medical procedures. Here, we illustrate how material sciences for clinical applications, critically depend on all aspects of biomedical sciences, including the field of epigenetics.
Collapse
Affiliation(s)
- Anne Krüger-Genge
- Department of Biomaterials and Healthcare, Fraunhofer-Institute for Applied Polymer Research (IAP), Division of Life Science and Bioprocesses, 14476 Potsdam-Golm, Germany.
| | - Olivia Mauger
- Department of Biomaterials and Healthcare, Fraunhofer-Institute for Applied Polymer Research (IAP), Division of Life Science and Bioprocesses, 14476 Potsdam-Golm, Germany.
| | - Joachim Storsberg
- Department of Biomaterials and Healthcare, Fraunhofer-Institute for Applied Polymer Research (IAP), Division of Life Science and Bioprocesses, 14476 Potsdam-Golm, Germany.
| | - Christian Schmidt
- Department of Biomaterials and Healthcare, Fraunhofer-Institute for Applied Polymer Research (IAP), Division of Life Science and Bioprocesses, 14476 Potsdam-Golm, Germany.
| |
Collapse
|