1
|
Chen X, Peng Y, Liu XS. DNA Methylation in Long-Term Memory. Physiology (Bethesda) 2025; 40:0. [PMID: 39907057 DOI: 10.1152/physiol.00032.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 01/06/2025] [Accepted: 01/30/2025] [Indexed: 02/06/2025] Open
Abstract
Understanding the neural mechanisms of memory has been one of the key questions in biology. Long-term memory, specifically, allows one to travel mentally without constraints of time and space. A long-term memory must have gone through a series of temporal processes: encoding, consolidation, storage, and retrieval. Decades of studies have revealed cellular and molecular mechanisms underlying each process. In this article, we first review the emerging concept of memory engrams and technologies of engram labeling, as these methods provide a new avenue to study the molecular mechanisms for memory. Then, we focus on DNA methylation and its role in long-term memory. Finally, we discuss some key remaining questions in this field and their implications in memory-related disease.
Collapse
Affiliation(s)
- Xinyue Chen
- Department of Neuroscience, Columbia University, New York, New York, United States
- Department of Pathology and Cell Biology, Columbia University Medical Center, Columbia University, New York, New York, United States
- Department of Neurology, Columbia University Medical Center, Columbia University, New York, New York, United States
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center, Columbia University, New York, New York, United States
| | - Yueqing Peng
- Department of Pathology and Cell Biology, Columbia University Medical Center, Columbia University, New York, New York, United States
- Department of Neurology, Columbia University Medical Center, Columbia University, New York, New York, United States
| | - X Shawn Liu
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center, Columbia University, New York, New York, United States
| |
Collapse
|
2
|
Kang YK, Min B, Eom J, Park JS, Jang J, Jeong S. Emergence of CpG-cluster blanket methylation in aged tissues: a novel signature of epigenomic aging. Nucleic Acids Res 2025; 53:gkaf354. [PMID: 40347138 PMCID: PMC12065108 DOI: 10.1093/nar/gkaf354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 03/20/2025] [Accepted: 05/07/2025] [Indexed: 05/12/2025] Open
Abstract
Aging is accompanied by widespread DNA methylation changes across the genome. While age-related methylation studies typically focus on individual CpGs, cluster analysis provides more robust data and improved interpretation. We characterized age-associated CpG-cluster methylation changes in mouse spleens, peripheral blood mononuclear cells, and livers. We identified a novel signature termed blanket methylations (BMs), fully methylated CpG clusters absent in young tissues but appearing in aged tissues. BM formation was locus- and cell-dependent, with minimal overlap among tissues. Statistical analysis, heterogeneity assessment, and random modeling demonstrated that BMs arise through nonrandom mechanisms and correlate with accelerated aging. Notably, BMs appeared in chronologically young mice with progeroid or disease-driven aging, including in 4-month-old Zmpste24-/- (lifespan ∼5 months) and 3-month-old Huntington's disease model mice (lifespan ∼4 months). The detection of BMs in purified CD4+ T cells demonstrated that their occurrence is intrinsic to aging cells rather than a result of infiltration from other tissues. Further investigation revealed age-related downregulation of zinc-finger-CxxC-domain genes, including Tet1 and Tet3, which protect CpG islands from methylation. Importantly, TET1 or TET3 depletion induced BM formation, linking their loss to age-associated methylation drift. These findings establish BMs as a robust marker of epigenomic aging, providing insight into age-related methylation changes.
Collapse
Affiliation(s)
- Yong-Kook Kang
- Aging Convergence Research Center (ACRC), Development and Differentiation Research Center, Korea Research Institute of Bioscience Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, South Korea
- Department of Functional Genomics, University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon 34113, South Korea
| | - Byungkuk Min
- Aging Convergence Research Center (ACRC), Development and Differentiation Research Center, Korea Research Institute of Bioscience Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, South Korea
| | - Jaemin Eom
- Aging Convergence Research Center (ACRC), Development and Differentiation Research Center, Korea Research Institute of Bioscience Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, South Korea
- Department of Functional Genomics, University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon 34113, South Korea
| | - Jung Sun Park
- Aging Convergence Research Center (ACRC), Development and Differentiation Research Center, Korea Research Institute of Bioscience Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, South Korea
| | - Jaewoong Jang
- Aging Convergence Research Center (ACRC), Development and Differentiation Research Center, Korea Research Institute of Bioscience Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, South Korea
| | - Sangkyun Jeong
- Genomics Department, Keyomics Co. Ltd, 17 Techno4-ro, Yuseong-gu, Daejeon 34013, South Korea
| |
Collapse
|
3
|
Zhao HG, Ren SK, Zhou WT, Liu ZM, Wang LM, Tian YJ, Zhao F. Immunohistochemical detection of 5-hydroxymethylcytosine as a prognostic biomarker in non-WNT/Non-SHH medulloblastomas. Sci Rep 2025; 15:15633. [PMID: 40325038 PMCID: PMC12053619 DOI: 10.1038/s41598-025-00052-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Accepted: 04/24/2025] [Indexed: 05/07/2025] Open
Abstract
Although reduced levels of global 5-hydroxymethylcytosine (5hmC) have been observed in medulloblastomas (MBs), it remains unclear whether immunohistochemical (IHC) evaluation of 5hmC can serve as a prognostic biomarker for patients with MB. We performed IHC staining using a 5hmC antibody on a cohort of 114 pediatric MBs, including 69 non-WNT/non-SHH MBs. The 5hmC staining score was evaluated using a 9-point scale based on both the staining intensity and the percentage positive cells. We found that a low 5hmC staining score (< 5 points) was associated with poor outcomes in patients with non-WNT/non-SHH MB (both P < 0.001). Multivariate Cox regression analyses demonstrated the 5hmC staining score was an independent prognostic predictor for progression-free survival (P < 0.001) and overall survival (P = 0.004) in patients with non-WNT/non-SHH MB. Both the receiver operating characteristic curves and calibration curves demonstrated the excellent performance of the nomogram models established based on the Cox regression models. The high predictive accuracy of the nomogram models was confirmed in a validation cohort comprising 32 patients with non-WNT/non-SHH MB. In conclusion, IHC evaluation of 5hmC may serve as a cost-effective and readily accessible approach for the prognostic stratification of patients with non-WNT/non-SHH MB.
Collapse
Affiliation(s)
- Han-Guang Zhao
- Department of Pediatric Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, 119 south 4th Ring West Road, Fengtai District, Beijing, 100070, China
| | - Si-Kang Ren
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Wen-Tao Zhou
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Cell Biology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Zhi-Ming Liu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Lei-Ming Wang
- Department of Pathology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yong-Ji Tian
- Department of Pediatric Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, 119 south 4th Ring West Road, Fengtai District, Beijing, 100070, China
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Fu Zhao
- Department of Pediatric Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, 119 south 4th Ring West Road, Fengtai District, Beijing, 100070, China.
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
4
|
Farmiloe G, Bejczy V, Tabolacci E, Willemsen R, Jacobs F. Transcriptomic profiling of unmethylated full mutation carriers implicates TET3 in FMR1 CGG repeat expansion methylation dynamics in fragile X syndrome. J Neurodev Disord 2025; 17:22. [PMID: 40287634 PMCID: PMC12032669 DOI: 10.1186/s11689-025-09609-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 04/01/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND Fragile X syndrome (FXS) is a neurodevelopmental disorder caused by the expansion of a CGG repeat in the 5'UTR of the FMR1 (fragile X messenger ribonucleoprotein 1) gene. Healthy individuals possess a repeat 30-55 CGG units in length. Once the CGG repeat exceeds 200 copies it triggers methylation at the locus. This methylation covers the FMR1 promoter region and silences expression of the gene and the production of FMRP (fragile X messenger ribonucleoprotein). The loss of FMRP is responsible for a number of pathologies including neurodevelopmental delay and autism spectrum disorder. Methylation of the expanded repeat in the FMR1 locus is the causal factor for FXS, however it is not known why the expanded repeat triggers this epigenetic change or how exactly DNA methylation is established. Intriguingly, genetic engineering of expanded CGG repeats of over 300 copies in the FMR1 locus in mice remains unmethylated. Also in humans, in very rare cases, individuals can have an FMR1 CGG expansion > 200 copies but the locus remains unmethylated. These unmethylated full mutation (UFM) individuals give us a rare opportunity to investigate the mechanism of FMR1 promoter methylation. METHODS Fibroblasts were obtained from a healthy control, an FXS patient and two unmethylated full expansion carriers. RNA was extracted and comparative transcriptomic analysis was performed on all samples. Whole genome sequencing was carried out on DNA from the two UFM carriers and the results analysed to investigate DNA variants that could explain the observed differences in gene expression. RESULTS Our analyses focused on genes involved in epigenetic modification. We show that Tet methylcytosine dioxygenase 3 (TET3), a gene involved in DNA methylation, is significantly downregulated in UFM carriers compared to healthy controls or FXS patient derived cells. Genomic analyses reveal a number of rare variants present in the TET3 locus in UFM carriers when compared to the reference genome. However, no clear modifying TET3 variants were identified. CONCLUSION Our results suggest that TET3 is a candidate factor responsible for the lack of methylation of the expanded FMR1 locus. Further analyses are needed to further elucidate this relationship, however given its potential to directly interact with CGG repeats and its ambiguous role in 5-hydroxy-methylation of CG containing sequences, TET3 is a strong candidate for further exploration.
Collapse
Affiliation(s)
- Grace Farmiloe
- Evolutionary Neurogenomics, University of Amsterdam, Swammerdam Institute for Life Sciences, Science Park 904, Amsterdam, 1098XH, The Netherlands
| | - Veronika Bejczy
- Evolutionary Neurogenomics, University of Amsterdam, Swammerdam Institute for Life Sciences, Science Park 904, Amsterdam, 1098XH, The Netherlands
| | - Elisabetta Tabolacci
- Department of Life Sciences and Public Health, Section of Genomic Medicine, Catholic University, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Rob Willemsen
- CBG-Department of Clinical Genetics, Erasmus MC, Rotterdam, The Netherlands
| | - Frank Jacobs
- Evolutionary Neurogenomics, University of Amsterdam, Swammerdam Institute for Life Sciences, Science Park 904, Amsterdam, 1098XH, The Netherlands.
- Amsterdam Neuroscience, Amsterdam, The Netherlands.
| |
Collapse
|
5
|
Liu Y, Wang XQ, Zhang P, Haghparast A, He WB, Zhang JJ. Research progress of DNA methylation on the regulation of substance use disorders and the mechanisms. Front Cell Neurosci 2025; 19:1566001. [PMID: 40230379 PMCID: PMC11994631 DOI: 10.3389/fncel.2025.1566001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Accepted: 03/17/2025] [Indexed: 04/16/2025] Open
Abstract
Drug abuse can damage the central nervous system and lead to substance use disorder (SUD). SUD is influenced by both genetic and environmental factors. Genes determine an individual's susceptibility to drug, while the dysregulation of epigenome drives the abnormal transcription processes, promoting the development of SUD. One of the most widely studied epigenetic mechanisms is DNA methylation, which can be inherited stably. In ontogeny, DNA methylation pattern is dynamic. DNA dysmethylation is prevalent in drug-related psychiatric disorders, resulting in local hypermethylation and transcriptional silencing of related genes. In this review, we summarize the role and regulatory mechanisms of DNA methylation in cocaine, opioids, and methamphetamine in terms of drug exposure, addiction memory, withdrawal relapse, intergenerational inheritance, and focus on cell-specific aspects of the studies with a view to suggesting possible therapeutic regimens for targeting methylation in both human and animal research.
Collapse
Affiliation(s)
- Ya Liu
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, National International Joint Research Center for Molecular Chinese Medicine, Shanxi University of Chinese Medicine, Jinzhong, China
| | - Xiao-Qian Wang
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, National International Joint Research Center for Molecular Chinese Medicine, Shanxi University of Chinese Medicine, Jinzhong, China
| | - Peng Zhang
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, National International Joint Research Center for Molecular Chinese Medicine, Shanxi University of Chinese Medicine, Jinzhong, China
| | - Abbas Haghparast
- Neuroscience Research Center, Institute of Neuroscience and Cognition, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Wen-Bin He
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, National International Joint Research Center for Molecular Chinese Medicine, Shanxi University of Chinese Medicine, Jinzhong, China
| | - Jian-Jun Zhang
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, National International Joint Research Center for Molecular Chinese Medicine, Shanxi University of Chinese Medicine, Jinzhong, China
| |
Collapse
|
6
|
Zhao Z, Chen S, Liu Z, Su J, Lü J, Hao L, Dou Y, Wang L, Song S. T7 Endonuclease I-Mediated Single-Base Mismatch Biosensing Strategy for High-Resolution Quantitative Analysis of 5-Hydroxymethylcytosine in Genomic DNA. JACS AU 2025; 5:1320-1327. [PMID: 40151246 PMCID: PMC11937965 DOI: 10.1021/jacsau.4c01184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 01/15/2025] [Accepted: 01/16/2025] [Indexed: 03/29/2025]
Abstract
5-hydroxymethylcytosine (5hmC) plays a pivotal role in the DNA demethylation pathway and transcriptional regulation. While sequencing-based methods such as TET-assisted bisulfite sequencing offer single-base resolution, they are not ideal for dynamic, time-sensitive quantification. Here, we present a novel enzymatic biosensing strategy leveraging T7 endonuclease I for rapid and locus-specific 5hmC detection with a single-base resolution. This electrochemical platform captures double-tagged dsDNA and detects 5hmC by monitoring the signal reduction upon T7 endonuclease cleavage of A-C mismatches. The method achieved high sensitivity, detecting as little as 10 pg of hydroxymethylated DNA amid a 100,000-fold excess of methylated or unmethylated DNA. Furthermore, we demonstrated its ability to quantify real-time 5hmC variation during umbilical cord mesenchymal stem cell differentiation. This approach offers a powerful tool for 5hmC analysis in dynamic biological processes.
Collapse
Affiliation(s)
- Zhihan Zhao
- Institute
of Materiobiology, College of Science, Shanghai
University, Shanghai 200444, China
- Shanghai
Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China
| | - Shixing Chen
- Shanghai
Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China
| | - Zhixiao Liu
- Department
of Histology and Embryology, Naval Medical
University, Shanghai 200433, China
| | - Jing Su
- Shanghai
Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China
| | - Junhong Lü
- School
of Pharmacy, Binzhou Medical University, Yantai 264003, China
| | - Lihong Hao
- Ningbo
Junkang Medical Technology Co., Ltd., Ningbo 315000, China
| | - Yanzhi Dou
- Shanghai
Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China
- Science and
Technology on Microsystem Laboratory, Shanghai
Institute of Microsystem and Information Technology Chinese Academy
of Science, Shanghai 200050, China
| | - Lihua Wang
- Institute
of Materiobiology, College of Science, Shanghai
University, Shanghai 200444, China
| | - Shiping Song
- Institute
of Materiobiology, College of Science, Shanghai
University, Shanghai 200444, China
- Shanghai
Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China
| |
Collapse
|
7
|
Zhao J, Gu T, Gao C, Miao G, Palma-Gudiel H, Yu L, Yang J, Wang Y, Li Y, Lim J, Li R, Yao B, Wu H, Schneider JA, Seyfried N, Grodstein F, De Jager PL, Jin P, Bennett DA. Brain 5-hydroxymethylcytosine alterations are associated with Alzheimer's disease neuropathology. Nat Commun 2025; 16:2842. [PMID: 40121201 PMCID: PMC11929800 DOI: 10.1038/s41467-025-58159-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 03/11/2025] [Indexed: 03/25/2025] Open
Abstract
5-hydroxymethylcytosine, also known as the sixth DNA base of the genome, plays an important role in brain aging and neurological disorders such as Alzheimer's disease. However, little is known about its genome-wide distribution and its association with Alzheimer's disease pathology. Here, we report a genome-wide profiling of 5-hydroxymethylcytosine in 1079 autopsied brains (dorsolateral prefrontal cortex) of older individuals and assess its association with multiple measures of Alzheimer's disease pathologies, including pathological diagnosis of Alzheimer's disease, amyloid-β load, and PHFtau tangle density. Of 197,765 5-hydroxymethylcytosine regions detected, we identified 2821 differentially hydroxymethylated regions associated with Alzheimer's disease neuropathology after controlling for multiple testing and covariates. Many differentially hydroxymethylated regions are located within known Alzheimer's disease loci, such as RIN3, PLCG2, ITGA2B, and USP6NL. Integrative multi-omics analyses support a potential mechanistic role of 5-hydroxymethylcytosine alterations in Alzheimer's disease. Our study presents a large-scale genome-wide atlas of 5-hydroxymethylcytosine in Alzheimer's brain and offers insight into the mechanism underlying Alzheimer's disease pathogenesis.
Collapse
Affiliation(s)
- Jinying Zhao
- Health Informatics Institute, University of South Florida, Tampa, FL, USA.
| | - Tongjun Gu
- Department of Epidemiology, College of Public Health and Health Professions, University of Florida, Gainesville, FL, USA
| | - Cheng Gao
- Health Informatics Institute, University of South Florida, Tampa, FL, USA
| | - Guanhong Miao
- Health Informatics Institute, University of South Florida, Tampa, FL, USA
| | - Helena Palma-Gudiel
- Department of Epidemiology, College of Public Health and Health Professions, University of Florida, Gainesville, FL, USA
| | - Lei Yu
- Rush Alzheimer's Disease Center & Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Jingyun Yang
- Rush Alzheimer's Disease Center & Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Yanling Wang
- Rush Alzheimer's Disease Center & Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Yujing Li
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Junghwa Lim
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Ronghua Li
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Bing Yao
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Hao Wu
- Department of Biostatistics and Bioinformatics, Emory University School of Public Health, Atlanta, GA, USA
| | - Julie A Schneider
- Rush Alzheimer's Disease Center & Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Nicholas Seyfried
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Francine Grodstein
- Rush Alzheimer's Disease Center & Department of Internal Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Philip L De Jager
- Center for Translational & Computational Neuroimmunology, Department of Neurology and the Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York, NY, USA
| | - Peng Jin
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA.
| | - David A Bennett
- Rush Alzheimer's Disease Center & Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA.
| |
Collapse
|
8
|
Liu XD, Ye CC, Wang Y, Zhang XS, Wei HX, Xie LJ, Xie JX, Xu YR, Zhong LY, Li SH, Li XJ, Lin L. DNA methylation confers a cerebellum-specific identity in non-human primates. Zool Res 2025; 46:414-428. [PMID: 40091535 PMCID: PMC12000133 DOI: 10.24272/j.issn.2095-8137.2024.325] [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: 09/26/2024] [Accepted: 12/17/2024] [Indexed: 03/19/2025] Open
Abstract
Selective regulation of gene expression across distinct brain regions is crucial for establishing and maintaining subdivision identities. DNA methylation, a key regulator of gene transcription, modulates transcriptional activity through the conversion of 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC). While DNA methylation is hypothesized to play an essential role in shaping brain identity by influencing gene expression patterns, its direct contribution, especially in primates, remains largely unexplored. This study examined DNA methylation landscapes and transcriptional profiles across four brain regions, including the cortex, cerebellum, striatum, and hippocampus, using samples from 12 rhesus monkeys. The cerebellum exhibited distinct epigenetic and transcriptional signatures, with differentially methylated regions (DMRs) significantly enriched in metabolic pathways. Notably, genes harboring clustered differentially hydroxymethylated regions (DhMRs) overlapped with those implicated in schizophrenia. Moreover, 5mC located 1 kb upstream of the ATG start codon was correlated with gene expression and exhibited region-specific associations with 5hmC. These findings provide insights into the coordinated regulation of cerebellum-specific 5mC and 5hmC , highlighting their potential roles in defining cerebellar identity and contributing to neuropsychiatric diseases.
Collapse
Affiliation(s)
- Xiao-Dong Liu
- Guangdong Key Laboratory of Non-human Primate Research, Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangzhou, Guangdong 510632, China
| | - Chang-Cheng Ye
- Guangdong Key Laboratory of Non-human Primate Research, Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangzhou, Guangdong 510632, China
| | - Yang Wang
- Guangdong Key Laboratory of Non-human Primate Research, Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangzhou, Guangdong 510632, China
| | - Xiao-Song Zhang
- Guangdong Key Laboratory of Non-human Primate Research, Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangzhou, Guangdong 510632, China
| | - Hui-Xian Wei
- Guangdong Key Laboratory of Non-human Primate Research, Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangzhou, Guangdong 510632, China
| | - Lei-Jie Xie
- Guangdong Key Laboratory of Non-human Primate Research, Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangzhou, Guangdong 510632, China
| | - Jia-Xiang Xie
- Guangdong Key Laboratory of Non-human Primate Research, Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangzhou, Guangdong 510632, China
| | - Yan-Ru Xu
- Guangdong Key Laboratory of Non-human Primate Research, Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangzhou, Guangdong 510632, China
| | - Li-Ying Zhong
- Guangdong Key Laboratory of Non-human Primate Research, Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangzhou, Guangdong 510632, China
| | - Shi-Hua Li
- Guangdong Key Laboratory of Non-human Primate Research, Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangzhou, Guangdong 510632, China
| | - Xiao-Jiang Li
- Guangdong Key Laboratory of Non-human Primate Research, Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangzhou, Guangdong 510632, China
| | - Li Lin
- Guangdong Key Laboratory of Non-human Primate Research, Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangzhou, Guangdong 510632, China. E-mail:
| |
Collapse
|
9
|
Šimelis K, Belle R, Kawamura A. Unravelling 2-oxoglutarate turnover and substrate oxidation dynamics in 5-methylcytosine-oxidising TET enzymes. Commun Chem 2024; 7:305. [PMID: 39706884 DOI: 10.1038/s42004-024-01382-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 11/28/2024] [Indexed: 12/23/2024] Open
Abstract
Fe(II)- and 2-oxoglutarate (2OG)-dependent dioxygenases use 2OG and O2 cofactors to catalyse substrate oxidation and yield oxidised product, succinate, and CO2. Simultaneous detection of substrate and cofactors is difficult, contributing to a poor understanding of the dynamics between substrate oxidation and 2OG decarboxylation activities. Here, we profile 5-methylcytosine (5mC)-oxidising Ten-Eleven Translocation (TET) enzymes using MS and 1H NMR spectroscopy methods and reveal a high degree of substrate oxidation-independent 2OG turnover under a range of conditions. 2OG decarboxylase activity is substantial (>20% 2OG turned over after 1 h) in the absence of substrate, while, under substrate-saturating conditions, half of total 2OG consumption is uncoupled from substrate oxidation. 2OG kinetics are affected by substrate and non-substrate DNA oligomers, and the sequence-agnostic effects are observed in amoeboflagellate Naegleria gruberi NgTet1 and human TET2. TET inhibitors also alter uncoupled 2OG kinetics, highlighting the potential effect of 2OG dioxygenase inhibitors on the intracellular balance of 2OG/succinate.
Collapse
Affiliation(s)
- Klemensas Šimelis
- Chemistry Research Laboratory, Department of Chemistry, University of Oxford, Oxford, UK
| | - Roman Belle
- Chemistry-School of Natural and Environmental Sciences, Bedson Building, Newcastle University, Newcastle upon Tyne, UK
| | - Akane Kawamura
- Chemistry Research Laboratory, Department of Chemistry, University of Oxford, Oxford, UK.
- Chemistry-School of Natural and Environmental Sciences, Bedson Building, Newcastle University, Newcastle upon Tyne, UK.
| |
Collapse
|
10
|
Zhang Y, Peng Z, Guo M, Wang Y, Liu J, Liu Y, Li M, Wei T, Li P, Zhao Y, Wang Y. TET3-facilitated differentiation of human umbilical cord mesenchymal stem cells into oligodendrocyte precursor cells for spinal cord injury recovery. J Transl Med 2024; 22:1118. [PMID: 39707356 DOI: 10.1186/s12967-024-05929-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 11/30/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND Spinal cord injury (SCI) inflicts a severe burden on patients and lacks effective treatments. Owing to the poor regenerative capabilities of endogenous oligodendrocyte precursor cells (OPCs) following SCI, there is a growing interest in alternative sources, such as human umbilical cord mesenchymal stem cells (HUCMSCs). TET3 is a key DNA demethylase that plays an important role in neural differentiation, but its role in OPC formation is not well understood. This study aimed to explore the TET3-mediated one-step induction of HUCMSCs into OPCs. METHODS In vitro, HUCMSCs were induced into OPCs following TET3 overexpression. Changes of methylation and hydroxymethylation during differentiation were monitored, mechanisms involved in the TET3-driven HUCMSC differentiation into OPCs were identified by RNA sequencing. Methylation levels in NG2 and PDGFRA promoter region were detected using Bisulfite Polymerase Chain Reaction (BSP).In vivo, therapeutic effects of iOPCs were evaluated through a rat Allen's SCI model. RESULTS The in vitro analysis confirmed that TET3 enhances HUCMSC differentiation into OPCs, validitied by specific marker expression. The induced OPCs (iOPCs) exhibited methylation and hydroxymethylation patterns similar to native OPCs. BSP analysis demonstrated that TET3 overexpression significantly reduced CpG island methylation in the NG2 and PDGFRA promoter regions. RNA sequencing revealed that TET3 induces iOPCs to express a series of genes essential for OPC formation while inhibiting the signaling pathways that hinder OPC development. In a rat model of SCI, TET3-overexpressing HUCMSCs appear to have the potential to differentiate into iOPCs in vivo, suppressed secondary injury, and promoted functional recovery. The therapeutic effects of iOPCs on SCI were superior to those of standard mesenchymal stem cell treatments. CONCLUSIONS Our study demonstrated that TET3-mediated demethylation reshapes the methylation patterns of HUCMSCs, enabling their efficient one-step conversion into OPCs and significantly reducing the time required for cell preparation. This approach offers a potential strategy for early intervention in SCI. In an SCI model, TET3-induced OPCs contributed to spinal cord repair, providing novel insights into cell therapy strategies for SCI through the lens of methylation regulation.
Collapse
Affiliation(s)
- Yubo Zhang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Harbin Medical University, 2075 Qunli Seventh Avenue, Daoli District, Harbin, 150001, Heilongjiang Province, China
| | - Zhibin Peng
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Harbin Medical University, 2075 Qunli Seventh Avenue, Daoli District, Harbin, 150001, Heilongjiang Province, China
| | - Man Guo
- Department of Obstetrics, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, Heilongjiang, China
| | - Yangyang Wang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Harbin Medical University, 2075 Qunli Seventh Avenue, Daoli District, Harbin, 150001, Heilongjiang Province, China
| | - Jingsong Liu
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Harbin Medical University, 2075 Qunli Seventh Avenue, Daoli District, Harbin, 150001, Heilongjiang Province, China
| | - Yishu Liu
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Harbin Medical University, 2075 Qunli Seventh Avenue, Daoli District, Harbin, 150001, Heilongjiang Province, China
| | - Mi Li
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Harbin Medical University, 2075 Qunli Seventh Avenue, Daoli District, Harbin, 150001, Heilongjiang Province, China
| | - Tianli Wei
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Harbin Medical University, 2075 Qunli Seventh Avenue, Daoli District, Harbin, 150001, Heilongjiang Province, China
| | - Pengfei Li
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Harbin Medical University, 2075 Qunli Seventh Avenue, Daoli District, Harbin, 150001, Heilongjiang Province, China
| | - Yingwei Zhao
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Harbin Medical University, 2075 Qunli Seventh Avenue, Daoli District, Harbin, 150001, Heilongjiang Province, China
| | - Yansong Wang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Harbin Medical University, 2075 Qunli Seventh Avenue, Daoli District, Harbin, 150001, Heilongjiang Province, China.
| |
Collapse
|
11
|
Ren X, Yang Y, Wang M, Yuan Q, Suo N, Xie X. Vitamin C and MEK Inhibitor PD0325901 Synergistically Promote Oligodendrocytes Generation by Promoting DNA Demethylation. Molecules 2024; 29:5939. [PMID: 39770028 PMCID: PMC11677943 DOI: 10.3390/molecules29245939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 12/06/2024] [Accepted: 12/14/2024] [Indexed: 01/11/2025] Open
Abstract
DNA methylation and demethylation are key epigenetic events that regulate gene expression and cell fate. DNA demethylation via oxidation of 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC) is typically mediated by TET (ten-eleven translocation) enzymes. The 5hmC modification is considered an intermediate state of DNA demethylation; it is particularly prevalent in the brain and is believed to play a role in the development of many cell types in the brain. Our previous studies have identified that vitamin C (Vc) and MEK inhibitor PD0325901 could significantly promote OPC (oligodendrocyte progenitor cell)-to-OL (oligodendrocyte) differentiation. Here we discovered that Vc and PD0325901 may promote OPC-to-OL differentiation by inducing DNA demethylation via hydroxymethylation. Blocking 5hmC formation almost totally blocked Vc- and PD0325901-stimulated OPC-to-OL differentiation. In addition, TET1 is not involved in Vc,- and PD0325901-promoted OL generation. We also found a synergistic effect between the two compounds in inducing OL generation, suggesting the possibility of a combination therapy for demyelination diseases in the future.
Collapse
Affiliation(s)
- Xinyue Ren
- State Key Laboratory of Drug Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; (X.R.); (Y.Y.); (M.W.); (Q.Y.)
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ying Yang
- State Key Laboratory of Drug Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; (X.R.); (Y.Y.); (M.W.); (Q.Y.)
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Min Wang
- State Key Laboratory of Drug Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; (X.R.); (Y.Y.); (M.W.); (Q.Y.)
| | - Qianting Yuan
- State Key Laboratory of Drug Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; (X.R.); (Y.Y.); (M.W.); (Q.Y.)
| | - Na Suo
- State Key Laboratory of Drug Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; (X.R.); (Y.Y.); (M.W.); (Q.Y.)
| | - Xin Xie
- State Key Laboratory of Drug Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; (X.R.); (Y.Y.); (M.W.); (Q.Y.)
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| |
Collapse
|
12
|
Gillespie CA, Chowdhury A, Quinn KA, Jenkins MW, Rollins AM, Watanabe M, Ford SM. Fundamentals of DNA methylation in development. Pediatr Res 2024:10.1038/s41390-024-03674-7. [PMID: 39658604 DOI: 10.1038/s41390-024-03674-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 09/12/2024] [Accepted: 09/30/2024] [Indexed: 12/12/2024]
Abstract
DNA methyation is critical to regulation of gene expression especially during developmentally dynamic changes. A large proportion occurs at CpG (a cytosine followed by a guanine nucleotide) sites and impacts gene expression based on location, timing and level of DNA methylation. The spectrum of effects produced by DNA methylation ranges from inhibition to enhancement of gene expression. Here basic terms and concepts in the study of DNA methylation are introduced. In addition, some of the commonly used techniques to assay DNA methylation are explained. New methods that allow the precise addition and removal of DNA methylation at specific sites will likely enhance our understanding of DNA methylation in development and may even lead to long-lasting therapeutic strategies to cure diseases. IMPACT: Fundamentals of DNA methylation including its significance are made accessible to a broad audience. Common assays for detecting DNA methylation are explained succinctly. Developmental patterns of DNA methylation detected in commonly used animal models are discussed and explained. Novel methodologies to investigate consequences of DNA methylation and demethylation are introduced.
Collapse
Affiliation(s)
- Caitlyn A Gillespie
- Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Amrin Chowdhury
- Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Katie A Quinn
- Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Michael W Jenkins
- Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Andrew M Rollins
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Michiko Watanabe
- Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Stephanie M Ford
- Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA.
- Divisions of Neonatology and Pediatric Cardiology, UH Rainbow Babies and Children's Hospital, Cleveland, OH, 44106, USA.
| |
Collapse
|
13
|
Foong YH, Caldwell B, Thorvaldsen JL, Krapp C, Mesaros CA, Zhou W, Kohli RM, Bartolomei MS. TET1 displays catalytic and non-catalytic functions in the adult mouse cortex. Epigenetics 2024; 19:2374979. [PMID: 38970823 PMCID: PMC11229741 DOI: 10.1080/15592294.2024.2374979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 06/26/2024] [Indexed: 07/08/2024] Open
Abstract
TET1/2/3 dioxygenases iteratively demethylate 5-methylcytosine, beginning with the formation of 5-hydroxymethylcytosine (5hmC). The post-mitotic brain maintains higher levels of 5hmC than most peripheral tissues, and TET1 ablation studies have underscored the critical role of TET1 in brain physiology. However, deletion of Tet1 precludes the disentangling of the catalytic and non-catalytic functions of TET1. Here, we dissect these functions of TET1 by comparing adult cortex of Tet1 wildtype (Tet1 WT), a novel Tet1 catalytically dead mutant (Tet1 HxD), and Tet1 knockout (Tet1 KO) mice. Using DNA methylation array, we uncover that Tet1 HxD and KO mutations perturb the methylation status of distinct subsets of CpG sites. Gene ontology (GO) analysis on specific differential 5hmC regions indicates that TET1's catalytic activity is linked to neuronal-specific functions. RNA-Seq further shows that Tet1 mutations predominantly impact the genes that are associated with alternative splicing. Lastly, we performed High-performance Liquid Chromatography Mass-Spectrometry lipidomics on WT and mutant cortices and uncover accumulation of lysophospholipids lysophosphatidylethanolamine and lysophosphatidylcholine in Tet1 HxD cortex. In summary, we show that Tet1 HxD does not completely phenocopy Tet1 KO, providing evidence that TET1 modulates distinct cortical functions through its catalytic and non-catalytic roles.
Collapse
Affiliation(s)
- Yee Hoon Foong
- Department of Cell and Developmental Biology, Perelman School of Medicine, Smilow Center for Translational Research, Philadelphia, PA, USA
| | - Blake Caldwell
- Department of Cell and Developmental Biology, Perelman School of Medicine, Smilow Center for Translational Research, Philadelphia, PA, USA
- Department of Biological Sciences, Virginia Tech, Blacksburg, Virginia, USA
| | - Joanne L. Thorvaldsen
- Department of Cell and Developmental Biology, Perelman School of Medicine, Smilow Center for Translational Research, Philadelphia, PA, USA
| | - Christopher Krapp
- Department of Cell and Developmental Biology, Perelman School of Medicine, Smilow Center for Translational Research, Philadelphia, PA, USA
| | - Clementina A. Mesaros
- Translational Biomarkers Core, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center of Excellence in Environmental Toxicology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Wanding Zhou
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Children’s Hospital of Philadelphia (CHOP), University of Pennsylvania, Philadelphia, PA, USA
- Penn Epigenetics Institute, Smilow Center for Translational Rsearch, Philadelphia, PA, USA
| | - Rahul M. Kohli
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn Epigenetics Institute, Smilow Center for Translational Rsearch, Philadelphia, PA, USA
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Marisa S. Bartolomei
- Department of Cell and Developmental Biology, Perelman School of Medicine, Smilow Center for Translational Research, Philadelphia, PA, USA
- Penn Epigenetics Institute, Smilow Center for Translational Rsearch, Philadelphia, PA, USA
| |
Collapse
|
14
|
Mortillo M, Kennedy EG, Hermetz KM, Burt AA, Marsit CJ. Epigenetic landscape of 5-hydroxymethylcytosine and associations with gene expression in placenta. Epigenetics 2024; 19:2326869. [PMID: 38507502 PMCID: PMC10956631 DOI: 10.1080/15592294.2024.2326869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 03/01/2024] [Indexed: 03/22/2024] Open
Abstract
5-hydroxymethylcystosine (5hmC), is an intermediate product in the DNA demethylation pathway, but may act as a functional epigenetic modification. We have conducted the largest study of site-specific 5hmC in placenta to date using parallel bisulphite and oxidative bisulphite modification with array-based assessment. Incorporating parallel RNA-sequencing data allowed us to assess associations between 5hmC and gene expression, using expression quantitative trait hydroxymethylation (eQTHM) analysis. We identified ~ 47,000 loci with consistently elevated (systematic) 5hmC proportions. Systematic 5hmC was significantly depleted (p < 0.0001) at CpG islands (CGI), and enriched (p < 0.0001) in 'open sea' regions (CpG >4 kb from CGI). 5hmC was most and least abundant at CpGs in enhancers and active transcription start sites (TSS), respectively (p < 0.05). We identified 499 significant (empirical-p <0.05) eQTHMs within 1 MB of the assayed gene. At most (75.4%) eQTHMs, the proportion of 5hmC was positively correlated with transcript abundance. eQTHMs were significantly enriched among enhancer CpGs and depleted among CpGs in active TSS (p < 0.05 for both). Finally, we identified 107 differentially hydroxymethylated regions (DHMRs, p < 0.05) across 100 genes. Our study provides insight into placental distribution of 5hmC, and sheds light on the functional capacity of this epigenetic modification in placenta.
Collapse
Affiliation(s)
- Michael Mortillo
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Elizabeth G. Kennedy
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Karen M. Hermetz
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Amber A. Burt
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Carmen J. Marsit
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| |
Collapse
|
15
|
Zheng K, Lyu Z, Chen J, Chen G. 5-Hydroxymethylcytosine: Far Beyond the Intermediate of DNA Demethylation. Int J Mol Sci 2024; 25:11780. [PMID: 39519332 PMCID: PMC11546248 DOI: 10.3390/ijms252111780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 10/18/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Epigenetics plays a pivotal role in regulating gene expression and cellular differentiation. DNA methylation, involving the addition of methyl groups to specific cytosine bases, is a well-known epigenetic modification. The recent discovery of 5-hydroxymethylcytosine (5hmC) has provided new insights into cytosine modifications. 5hmC, derived from the oxidation of 5-methylcytosine (5mC), serves as both an intermediate in demethylation and a stable chemical modification in the genome. In this comprehensive review, we summarize the recent research advancements regarding the functions of 5hmC in development and disease. We discuss its implications in gene expression regulation, cellular differentiation, and its potential role as a diagnostic and prognostic marker in various diseases. Additionally, we highlight the challenges associated with accurately detecting and quantifying 5hmC and present the latest methodologies employed for its detection. Understanding the functional role of 5hmC in epigenetic regulation and further advancing our understanding of gene expression dynamics and cellular processes hold immense promise for the development of novel therapeutic strategies and precision medicine approaches.
Collapse
Affiliation(s)
- Kaixi Zheng
- College of Life Sciences and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou 310018, China; (K.Z.); (Z.L.); (J.C.)
- School of Life Sciences, Central South University, Changsha 410031, China
| | - Zhengbing Lyu
- College of Life Sciences and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou 310018, China; (K.Z.); (Z.L.); (J.C.)
| | - Jianqing Chen
- College of Life Sciences and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou 310018, China; (K.Z.); (Z.L.); (J.C.)
| | - Guodong Chen
- School of Life Sciences, Central South University, Changsha 410031, China
| |
Collapse
|
16
|
Liu JW, Zhang ZQ, Zhu ZC, Li K, Xu Q, Zhang J, Cheng XW, Li H, Sun Y, Wang JJ, Hu LL, Xiong ZQ, Zhu Y. Loss of TET Activity in the Postnatal Mouse Brain Perturbs Synaptic Gene Expression and Impairs Cognitive Function. Neurosci Bull 2024; 40:1699-1712. [PMID: 39395911 DOI: 10.1007/s12264-024-01302-2] [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: 11/21/2023] [Accepted: 06/13/2024] [Indexed: 10/14/2024] Open
Abstract
Conversion of 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC) by ten-eleven translocation (TET) family proteins leads to the accumulation of 5hmC in the central nervous system; however, the role of 5hmC in the postnatal brain and how its levels and target genes are regulated by TETs remain elusive. We have generated mice that lack all three Tet genes specifically in postnatal excitatory neurons. These mice exhibit significantly reduced 5hmC levels, altered dendritic spine morphology within brain regions crucial for cognition, and substantially impaired spatial and associative memories. Transcriptome profiling combined with epigenetic mapping reveals that a subset of genes, which display changes in both 5hmC/5mC levels and expression patterns, are involved in synapse-related functions. Our findings provide insight into the role of postnatally accumulated 5hmC in the mouse brain and underscore the impact of 5hmC modification on the expression of genes essential for synapse development and function.
Collapse
Affiliation(s)
- Ji-Wei Liu
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Ze-Qiang Zhang
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhi-Chuan Zhu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 20031, China
| | - Kui Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 20031, China
- Lingang Laboratory, Shanghai, 201602, China
| | - Qiwu Xu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 20031, China
- Lingang Laboratory, Shanghai, 201602, China
| | - Jing Zhang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 20031, China
| | - Xue-Wen Cheng
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 20031, China
| | - Han Li
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Ying Sun
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Ji-Jun Wang
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Lu-Lu Hu
- Fudan University Institutes of Biomedical Sciences, Shanghai Cancer Center, Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Shanghai Medical College of Fudan University, Shanghai, 200032, China
| | - Zhi-Qi Xiong
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 20031, China.
- Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, 201602, China.
| | - Yongchuan Zhu
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
| |
Collapse
|
17
|
Stein RA, Gomaa FE, Raparla P, Riber L. Now and then in eukaryotic DNA methylation. Physiol Genomics 2024; 56:741-763. [PMID: 39250426 DOI: 10.1152/physiolgenomics.00091.2024] [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: 07/01/2024] [Accepted: 09/06/2024] [Indexed: 09/11/2024] Open
Abstract
Since the mid-1970s, increasingly innovative methods to detect DNA methylation provided detailed information about its distribution, functions, and dynamics. As a result, new concepts were formulated and older ones were revised, transforming our understanding of the associated biology and catalyzing unprecedented advances in biomedical research, drug development, anthropology, and evolutionary biology. In this review, we discuss a few of the most notable advances, which are intimately intertwined with the study of DNA methylation, with a particular emphasis on the past three decades. Examples of these strides include elucidating the intricacies of 5-methylcytosine (5-mC) oxidation, which are at the core of the reversibility of this epigenetic modification; the three-dimensional structural characterization of eukaryotic DNA methyltransferases, which offered insights into the mechanisms that explain several disease-associated mutations; a more in-depth understanding of DNA methylation in development and disease; the possibility to learn about the biology of extinct species; the development of epigenetic clocks and their use to interrogate aging and disease; and the emergence of epigenetic biomarkers and therapies.
Collapse
Affiliation(s)
- Richard A Stein
- Department of Chemical and Biomolecular Engineering, NYU Tandon School of Engineering, Brooklyn, New York, United States
| | - Faris E Gomaa
- Department of Chemical and Biomolecular Engineering, NYU Tandon School of Engineering, Brooklyn, New York, United States
| | - Pranaya Raparla
- Department of Chemical and Biomolecular Engineering, NYU Tandon School of Engineering, Brooklyn, New York, United States
| | - Leise Riber
- Department of Plant and Environmental Sciences, University of Copenhagen, Frederiksberg, Denmark
| |
Collapse
|
18
|
Kamei N, Day K, Guo W, Haus DL, Nguyen HX, Scarfone VM, Booher K, Jia XY, Cummings BJ, Anderson AJ. Injured inflammatory environment overrides the TET2 shaped epigenetic landscape of pluripotent stem cell derived human neural stem cells. Sci Rep 2024; 14:25186. [PMID: 39448736 PMCID: PMC11502794 DOI: 10.1038/s41598-024-75689-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 10/08/2024] [Indexed: 10/26/2024] Open
Abstract
Spinal cord injury creates an inflammatory microenvironment that regulates the capacity of transplanted human Neural Stem Cells (hNSC) to migrate, differentiate, and repair injury. Despite similarities in gene expression and markers detected by immunostaining, hNSC populations exhibit heterogeneous therapeutic potential. This heterogeneity derives in part from the epigenetic landscape in the hNSC genome, specifically methylation (5mC) and hydroxymethylation (5hmC) state, which may affect the response of transplanted hNSC in the injury microenvironment and thereby modulate repair capacity. We demonstrate a significant up-regulation of methylcytosine dioxygenase 2 gene (TET2) expression in undifferentiated hNSC derived from human embryonic stem cells (hES-NSC), and report that this is associated with hES-NSC competence for differentiation marker expression. TET2 protein catalyzes active demethylation and TET2 upregulation could be a signature of pluripotent exit, while shaping the epigenetic landscape in hES-NSC. We determine that the inflammatory environment overrides epigenetic programming in vitro and in vivo by directly modulating TET2 expression levels in hES-NSC to change cell fate. We also report the effect of cell fate and microenvironment on differential methylation 5mC/5hmC balance. Understanding how the activity of epigenetic modifiers changes within the transplantation niche in vivo is crucial for assessment of hES-NSC behavior for potential clinical applications.
Collapse
Affiliation(s)
- Noriko Kamei
- Sue & Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, 92697-1705, USA.
- Anatomy and Neurobiology, University of California, Irvine, Irvine, CA, 92697-4475, USA.
| | - Kenneth Day
- Zymo Research Corp, 17062 Murphy Ave, Irvine, CA, 92614, USA
- Vidium Animal Health, 7201 E Henkel Way Suite210, Scottsdale, AZ, 85255, USA
| | - Wei Guo
- Zymo Research Corp, 17062 Murphy Ave, Irvine, CA, 92614, USA
| | - Daniel L Haus
- Sue & Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, 92697-1705, USA
| | - Hal X Nguyen
- Sue & Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, 92697-1705, USA
| | - Vanessa M Scarfone
- Sue & Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, 92697-1705, USA
| | - Keith Booher
- Zymo Research Corp, 17062 Murphy Ave, Irvine, CA, 92614, USA
| | - Xi-Yu Jia
- Zymo Research Corp, 17062 Murphy Ave, Irvine, CA, 92614, USA
| | - Brian J Cummings
- Sue & Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, 92697-1705, USA.
| | - Aileen J Anderson
- Sue & Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, 92697-1705, USA.
| |
Collapse
|
19
|
Bahabry R, Hauser RM, Sánchez RG, Jago SS, Ianov L, Stuckey RJ, Parrish RR, Ver Hoef L, Lubin FD. Alterations in DNA 5-hydroxymethylation patterns in the hippocampus of an experimental model of chronic epilepsy. Neurobiol Dis 2024; 200:106638. [PMID: 39142613 DOI: 10.1016/j.nbd.2024.106638] [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: 10/17/2023] [Revised: 07/27/2024] [Accepted: 08/11/2024] [Indexed: 08/16/2024] Open
Abstract
Temporal lobe epilepsy (TLE) is a type of focal epilepsy characterized by spontaneous recurrent seizures originating from the hippocampus. The epigenetic reprogramming hypothesis of epileptogenesis suggests that the development of TLE is associated with alterations in gene transcription changes resulting in a hyperexcitable network in TLE. DNA 5-methylcytosine (5-mC) is an epigenetic mechanism that has been associated with chronic epilepsy. However, the contribution of 5-hydroxymethylcytosine (5-hmC), a product of 5-mC demethylation by the Ten-Eleven Translocation (TET) family proteins in chronic TLE is poorly understood. 5-hmC is abundant in the brain and acts as a stable epigenetic mark altering gene expression through several mechanisms. Here, we found that the levels of bulk DNA 5-hmC but not 5-mC were significantly reduced in the hippocampus of human TLE patients and in the kainic acid (KA) TLE rat model. Using 5-hmC hMeDIP-sequencing, we characterized 5-hmC distribution across the genome and found bidirectional regulation of 5-hmC at intergenic regions within gene bodies. We found that hypohydroxymethylated 5-hmC intergenic regions were associated with several epilepsy-related genes, including Gal, SV2, and Kcnj11 and hyperdroxymethylation 5-hmC intergenic regions were associated with Gad65, TLR4, and Bdnf gene expression. Mechanistically, Tet1 knockdown in the hippocampus was sufficient to decrease 5-hmC levels and increase seizure susceptibility following KA administration. In contrast, Tet1 overexpression in the hippocampus resulted in increased 5-hmC levels associated with improved seizure resiliency in response to KA. These findings suggest an important role for 5-hmC as an epigenetic regulator of epilepsy that can be manipulated to influence seizure outcomes.
Collapse
Affiliation(s)
- Rudhab Bahabry
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, United States of America.
| | - Rebecca M Hauser
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, United States of America.
| | - Richard G Sánchez
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, United States of America.
| | - Silvienne Sint Jago
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, United States of America.
| | - Lara Ianov
- Civitan International Research Center, University of Alabama at Birmingham, Birmingham, AL, United States of America.
| | - Remy J Stuckey
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, United States of America.
| | - R Ryley Parrish
- Department of Cell Biology and Physiology, Brigham Young University, Provo, UT, United States of America.
| | - Lawrence Ver Hoef
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, United States of America.
| | - Farah D Lubin
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, United States of America.
| |
Collapse
|
20
|
Shen Y, Zhu W, Li S, Zhang Z, Zhang J, Li M, Zheng W, Wang D, Zhong Y, Li M, Zheng H, Du J. Integrated analyses of 5 mC, 5hmC methylation and gene expression reveal pathology-associated AKT3 gene and potential biomarkers for Alzheimer's disease. J Psychiatr Res 2024; 178:367-377. [PMID: 39197298 DOI: 10.1016/j.jpsychires.2024.08.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 07/18/2024] [Accepted: 08/14/2024] [Indexed: 09/01/2024]
Abstract
AIMS 5 mC methylation and hydroxymethylation (5hmC) are associated with Alzheimer's disease (AD). However, previous studies were limited by the absence of a 5hmC calculation. This study aims to find AD associated predictors and potential therapeutic chemicals using bioinformatics approach integrating 5 mC, 5hmC, and expression changes, and an AD mouse model. METHODS Gene expression microarray and 5 mC and 5hmC sequencing datasets were downloaded from GEO repository. 142 AD and 52 normal entorhinal cortex specimens were enrolled. Data from oxidative bisulfite sequencing (oxBS)-treated samples, which represent only 5 mC, were used to calculate 5hmC level. Functional analyses, random forest supervised classification and methylation validation were applied. Potential chemicals were predicted by CMap. Morris water maze, Y maze and novel object recognition behavior tests were performed using FAD4T AD mice model. Cortex and hippocampus tissues were isolated for immunohistochemical staining. RESULTS C1QTNF5, UBD, ZFP106, NEDD1, AKT3, and MBP genes involving 13 promoter CpG sites with 5mc, 5hmC methylation and expression difference were identified. AKT3 and MBP were down-regulated in both patients and mouse model. Three CpG sites in AKT3 and MBP showed significant methylation difference on validation. FAD4T AD mice showed recession in brain functions and lower AKT3 expression in both cortex and hippocampus. Ten chemicals were predicted as potential treatments for AD. CONCLUSIONS AKT3 and MBP may be associated with AD pathology and could serve as biomarkers. The ten predicted chemicals might offer new therapeutic approaches. Our findings could contribute to identifying novel markers and advancing the understanding of AD mechanisms.
Collapse
Affiliation(s)
- Yupei Shen
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Weiqiang Zhu
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Shuaicheng Li
- School of Computer Science, Fudan University, Shanghai Key Laboratory of Intelligent Information Processing Shanghai, China
| | - Zhaofeng Zhang
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Jian Zhang
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Mingjie Li
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
| | - Wei Zheng
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Difei Wang
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Yushun Zhong
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Min Li
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Huajun Zheng
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Jing Du
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China.
| |
Collapse
|
21
|
Wang M, Wang L, Huang Y, Qiao Z, Yi S, Zhang W, Wang J, Yang G, Cui X, Kou X, Zhao Y, Wang H, Jiang C, Gao S, Chen J. Loss of Tet hydroxymethylase activity causes mouse embryonic stem cell differentiation bias and developmental defects. SCIENCE CHINA. LIFE SCIENCES 2024; 67:2132-2148. [PMID: 39037697 DOI: 10.1007/s11427-024-2631-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 05/23/2024] [Indexed: 07/23/2024]
Abstract
The TET family is well known for active DNA demethylation and plays important roles in regulating transcription, the epigenome and development. Nevertheless, previous studies using knockdown (KD) or knockout (KO) models to investigate the function of TET have faced challenges in distinguishing its enzymatic and nonenzymatic roles, as well as compensatory effects among TET family members, which has made the understanding of the enzymatic role of TET not accurate enough. To solve this problem, we successfully generated mice catalytically inactive for specific Tet members (Tetm/m). We observed that, compared with the reported KO mice, mutant mice exhibited distinct developmental defects, including growth retardation, sex imbalance, infertility, and perinatal lethality. Notably, Tetm/m mouse embryonic stem cells (mESCs) were successfully established but entered an impaired developmental program, demonstrating extended pluripotency and defects in ectodermal differentiation caused by abnormal DNA methylation. Intriguingly, Tet3, traditionally considered less critical for mESCs due to its lower expression level, had a significant impact on the global hydroxymethylation, gene expression, and differentiation potential of mESCs. Notably, there were common regulatory regions between Tet1 and Tet3 in pluripotency regulation. In summary, our study provides a more accurate reference for the functional mechanism of Tet hydroxymethylase activity in mouse development and ESC pluripotency regulation.
Collapse
Affiliation(s)
- Mengting Wang
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, 200092, China
| | - Liping Wang
- Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, 200092, China
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of the Ministry of Education, Orthopaedic Department of Tongji Hospital, Tongji University, Shanghai, 200065, China
| | - Yanxin Huang
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, 200092, China
| | - Zhibin Qiao
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, 200092, China
| | - Shanru Yi
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, 200092, China
| | - Weina Zhang
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, 200092, China
| | - Jing Wang
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, 200092, China
| | - Guang Yang
- Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, 200092, China
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of the Ministry of Education, Orthopaedic Department of Tongji Hospital, Tongji University, Shanghai, 200065, China
| | - Xinyu Cui
- Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, 200092, China
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of the Ministry of Education, Orthopaedic Department of Tongji Hospital, Tongji University, Shanghai, 200065, China
| | - Xiaochen Kou
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, 200092, China
| | - Yanhong Zhao
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, 200092, China
| | - Hong Wang
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, 200092, China
| | - Cizhong Jiang
- Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China.
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, 200092, China.
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of the Ministry of Education, Orthopaedic Department of Tongji Hospital, Tongji University, Shanghai, 200065, China.
| | - Shaorong Gao
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, 200092, China.
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, 200120, China.
| | - Jiayu Chen
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
22
|
Kong Y, Ji J, Zhan X, Yan W, Liu F, Ye P, Wang S, Tai J. Tet1-mediated 5hmC regulates hippocampal neuroinflammation via wnt signaling as a novel mechanism in obstructive sleep apnoea leads to cognitive deficit. J Neuroinflammation 2024; 21:208. [PMID: 39169375 PMCID: PMC11340128 DOI: 10.1186/s12974-024-03189-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 07/29/2024] [Indexed: 08/23/2024] Open
Abstract
BACKGROUND Obstructive sleep apnoea (OSA) is a sleep-disordered breathing characterized by intermittent hypoxia (IH) that may cause cognitive dysfunction. However, the impact of IH on molecular processes involved in cognitive function remains unclear. METHODS C57BL / 6 J mice were exposed to either normoxia (control) or IH for 6 weeks. DNA hydroxymethylation was quantified by hydroxymethylated DNA immunoprecipitation (hMeDIP) sequencing. ten-eleven translocation 1 (Tet1) was knocked down by lentivirus. Specifically, cognitive function was assessed by behavioral experiments, pathological features were assessed by HE staining, the hippocampal DNA hydroxymethylation was examined by DNA dot blot and immunohistochemical staining, while the Wnt signaling pathway and its downstream effects were studied using qRT-PCR, immunofluorescence staining, and Luminex liquid suspension chip analysis. RESULTS IH mice showed pathological changes and cognitive dysfunction in the hippocampus. Compared with the control group, IH mice exhibited global DNA hydroxylmethylation in the hippocampus, and the expression of three hydroxylmethylases increased significantly. The Wnt signaling pathway was activated, and the mRNA and 5hmC levels of Wnt3a, Ccnd2, and Prickle2 were significantly up-regulated. Further caused downstream neurogenesis abnormalities and neuroinflammatory activation, manifested as increased expression of IBA1 (a marker of microglia), GFAP (a marker of astrocytes), and DCX (a marker of immature neurons), as well as a range of inflammatory cytokines (e.g. TNFa, IL3, IL9, and IL17A). After Tet1 knocked down, the above indicators return to normal. CONCLUSION Activation of Wnt signaling pathway by hippocampal Tet1 is associated with cognitive dysfunction induced by IH.
Collapse
Affiliation(s)
- Yaru Kong
- Children's Hospital Capital Institute of Pediatrics, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100020, China
- Graduate School of Peking Union Medical College, Beijing, 100730, China
- Department of Otolaryngology, Head and Neck Surgery, Children's Hospital Capital Institute of Paediatrics, Beijing, 100020, China
| | - Jie Ji
- Department of Otolaryngology, Head and Neck Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Xiaojun Zhan
- Department of Otolaryngology, Head and Neck Surgery, Children's Hospital Capital Institute of Paediatrics, Beijing, 100020, China
| | - Weiheng Yan
- Children's Hospital Capital Institute of Pediatrics, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100020, China
- Graduate School of Peking Union Medical College, Beijing, 100730, China
- Department of Otolaryngology, Head and Neck Surgery, Children's Hospital Capital Institute of Paediatrics, Beijing, 100020, China
| | - Fan Liu
- Children's Hospital Capital Institute of Pediatrics, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100020, China
- Graduate School of Peking Union Medical College, Beijing, 100730, China
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Pengfei Ye
- Department of Otolaryngology, Head and Neck Surgery, Children's Hospital Capital Institute of Paediatrics, Beijing, 100020, China
| | - Shan Wang
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, 100020, China.
| | - Jun Tai
- Children's Hospital Capital Institute of Pediatrics, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100020, China.
- Department of Otolaryngology, Head and Neck Surgery, Children's Hospital Capital Institute of Paediatrics, Beijing, 100020, China.
| |
Collapse
|
23
|
Kestering-Ferreira É, Heberle BA, Sindermann Lumertz F, Gobira PH, Orso R, Grassi-Oliveira R, Viola TW. Sex differences in sensitivity to fentanyl effects in mice: Behavioral and molecular findings during late adolescence. Neurosci Lett 2024; 837:137898. [PMID: 39013536 DOI: 10.1016/j.neulet.2024.137898] [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: 03/04/2024] [Revised: 07/09/2024] [Accepted: 07/10/2024] [Indexed: 07/18/2024]
Abstract
PURPOSE Sex differences play a crucial role in understanding vulnerability to opioid addiction, yet there have been limited preclinical investigations of this effect during the transition from adolescence to adulthood. The present study compared the behaviors of male and female rodents in response to fentanyl treatment and targeted molecular correlates in the striatum and medial prefrontal cortex. MATERIALS AND METHODS Thirty adolescent C57BL/6J mice underwent a 1-week fentanyl treatment with an escalating dose. In addition to evaluating locomotor activity and anxiety-related parameters, we also assessed naloxone-induced fentanyl acute withdrawal jumps. We employed real-time quantitative PCR (qPCR) to assess overall gene expression of dopaminergic receptors (Drd1, Drd2, Drd4 and Drd5) and the μ-opioid receptor Oprm1. The levels of epigenetic base modifications including 5-methylcytosine (5mC) and 5-hydroxymethylcytosine (5hmC) were assessed on CpG islands of relevant genes. RESULTS Females had higher locomotor activity than males after chronic fentanyl treatment, and they exhibited higher fentanyl withdrawal jumping behavior induced by naloxone. Females also presented lower Drd4 gene expression and DNA methylation (5mC + 5hmC) in the striatum. We found that locomotor activity and fentanyl withdrawal jumps were negatively correlated with Drd4 methylation and gene expression in the striatum, respectively. CONCLUSIONS The findings suggested that female mice displayed heightened sensitivity to the effects of fentanyl treatment during the transition from adolescence to adulthood. This effect may be associated with molecular alterations related to the Drd4 gene.
Collapse
MESH Headings
- Animals
- Fentanyl/pharmacology
- Male
- Female
- Mice, Inbred C57BL
- Sex Characteristics
- Receptors, Opioid, mu/genetics
- Receptors, Opioid, mu/metabolism
- Mice
- DNA Methylation/drug effects
- Analgesics, Opioid/pharmacology
- Corpus Striatum/metabolism
- Corpus Striatum/drug effects
- Locomotion/drug effects
- Prefrontal Cortex/drug effects
- Prefrontal Cortex/metabolism
- Receptors, Dopamine/genetics
- Receptors, Dopamine/metabolism
- Naloxone/pharmacology
- Behavior, Animal/drug effects
- Substance Withdrawal Syndrome/genetics
- Substance Withdrawal Syndrome/metabolism
- Epigenesis, Genetic/drug effects
Collapse
Affiliation(s)
- Érika Kestering-Ferreira
- School of Medicine, Brain Institute of Rio Grande do Sul, Developmental Cognitive Neuroscience Lab, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| | | | - Francisco Sindermann Lumertz
- School of Medicine, Brain Institute of Rio Grande do Sul, Developmental Cognitive Neuroscience Lab, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Pedro Henrique Gobira
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Rodrigo Orso
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Rodrigo Grassi-Oliveira
- School of Medicine, Brain Institute of Rio Grande do Sul, Developmental Cognitive Neuroscience Lab, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil; Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Thiago Wendt Viola
- School of Medicine, Brain Institute of Rio Grande do Sul, Developmental Cognitive Neuroscience Lab, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil.
| |
Collapse
|
24
|
Choza JI, Virani M, Kuhn NC, Adams M, Kochmanski J, Bernstein AI. Parkinson's disease-associated shifts between DNA methylation and DNA hydroxymethylation in human brain in PD-related genes, including PARK19 (DNAJC6) and PTPRN2 (IA-2β). RESEARCH SQUARE 2024:rs.3.rs-4572401. [PMID: 39070644 PMCID: PMC11275970 DOI: 10.21203/rs.3.rs-4572401/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Background The majority of Parkinson's disease (PD) cases are due to a complex interaction between aging, genetics, and environmental factors; epigenetic mechanisms are thought to act as important mediators of these risk factors. While multiple studies to date have explored the role of DNA modifications in PD, few focus on 5-hydroxymethylcytosine (5hmC). Because 5hmC occurs at its highest levels in the brain and is thought to be particularly important in the central nervous system, particularly in the response to neurotoxicants, it is important to explore the potential role of 5hmC in PD. This study expands on our previously published epigenome-wide association study (EWAS) performed on DNA isolated from neuron-enriched nuclei from human postmortem parietal cortex from the Banner Sun Health Research Institute Brain Bank. The study aimed to identify paired changes in 5hmC and 5mC in PD in enriched neuronal nuclei isolated from PD post-mortem parietal cortex and age- and sex-matched controls. We performed oxidative bisulfite (oxBS) conversion and paired it with our previously published bisulfite (BS)-based EWAS on the same samples to identify cytosines with significant shifts between these two related epigenetic marks. Interaction differentially modified cytosines (iDMCs) were identified using our recently published mixed-effects model for co-analyzing βmC and βhmC data. Results We identified 1,030 iDMCs with paired changes in 5mC and 5hmC (FDR < 0.05) that map to 695 genes, including PARK19 (DNAJC6), a familial PD gene, and PTPRN2 (IA-2), which has been previously implicated in PD in both epigenetic and mechanistic studies. The majority of iDMC-containing genes have not previously been implicated in PD and were not identified in our previous BS-based EWAS. Conclusions These data potentially link epigenetic regulation of the PARK19 and PTPRN2 loci in the pathogenesis of idiopathic PD. In addition, iDMC-containing genes have known functions in synaptic formation and function, cell cycle and senescence, neuroinflammation, and epigenetic regulation. These data suggest that there are significant shifts between 5mC and 5hmC associated with PD in genes relevant to PD pathogenesis that are not captured by analyzing BS-based data alone or by analyzing each mark as a distinct dataset.
Collapse
|
25
|
Wang J, Zhang Y, Ma Y, Zhao S, Wang J, Chen H, Zhang J, Liu J. TET1 inhibits liver fibrosis by blocking hepatic stellate cell activation. J Gastroenterol Hepatol 2024; 39:1403-1412. [PMID: 38369780 DOI: 10.1111/jgh.16443] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 11/21/2023] [Accepted: 11/22/2023] [Indexed: 02/20/2024]
Abstract
Hepatic stellate cells (HSCs) are critical regulator contributing to the onset and progression of liver fibrosis. Chronic liver injury triggers HSCs to undergo vast changes and trans-differentiation into a myofibroblast HSCs, the mechanism remains to be elucidated. This study investigated that the involvement of hydroxymethylase TET1 (ten-eleven translocation 1) in HSC activation and liver fibrosis. It is revealed that TET1 levels were downregulated in the livers in mouse models of liver fibrosis and patients with cirrhosis, as well as activated HSCs in comparison to quiescent HSCs. In vitro data showed that the inhibition of TET1 promoted the activation HSC, whereas TET1 overexpression inhibited HSC activation. Moreover, TET1 could regulate KLF2 (Kruppel-like transcription factors) transcription by promoting hydroxymethylation of its promoter, which in turn suppressed the activation of HSCs. In vivo, it is confirmed that liver fibrosis was aggravated in Tet1 knockout mice after CCl4 injection, accompanied by excessive activation of primary stellate cells, in contrast to wild-type mice. In conclusion, we suggested that TET1 plays a significant role in HSC activation and liver fibrosis, which provides a promising target for anti-fibrotic therapies.
Collapse
Affiliation(s)
- Jingjie Wang
- Department of Gastroenterology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Yitong Zhang
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Yanyun Ma
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Fudan University, Shanghai, China
- Department of Anthropology and Human Genetics, School of Life Sciences, Fudan University, Shanghai, China
- Six-sector Industrial Research Institute, Fudan University, Shanghai, China
| | - Suhan Zhao
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Jiucun Wang
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Fudan University, Shanghai, China
- Department of Anthropology and Human Genetics, School of Life Sciences, Fudan University, Shanghai, China
| | - Hongtan Chen
- Department of Gastroenterology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jun Zhang
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Jie Liu
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai, China
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Fudan University, Shanghai, China
| |
Collapse
|
26
|
Scatturice LA, Vázquez N, Strobl-Mazzulla PH. miR-137 confers robustness to the territorial restriction of the neural plate border. Development 2024; 151:dev202344. [PMID: 38828854 DOI: 10.1242/dev.202344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 05/20/2024] [Indexed: 06/05/2024]
Abstract
The neural plate border (NPB) of vertebrate embryos is segregated from the neural plate (NP) and epidermal regions, and comprises an intermingled group of progenitors with multiple fate potential. Recent studies have shown that, during the gastrula stage, TFAP2A acts as a pioneer factor in remodeling the epigenetic landscape required to activate components of the NPB induction program. Here, we show that chick Tfap2a has two highly conserved binding sites for miR-137, and both display a reciprocal expression pattern at the NPB and NP, respectively. In addition, ectopic miR-137 expression reduced TFAP2A, whereas its functional inhibition expanded their territorial distribution overlapping with PAX7. Furthermore, we demonstrate that loss of the de novo DNA methyltransferase DNMT3A expanded miR-137 expression to the NPB. Bisulfite sequencing revealed a markedly elevated presence of non-canonical CpH methylation within the miR-137 promoter region when comparing NPB and NP samples. Our findings show that miR-137 contributes to the robustness of NPB territorial restriction in vertebrate development.
Collapse
Affiliation(s)
- Luciana A Scatturice
- Laboratory of Developmental Biology, Instituto Tecnológico de Chascomús (CONICET-UNSAM). Escuela de Bio y Nanotecnologías (UNSAM), Chascomús, Buenos Aires 7130, Argentina
| | - Nicolás Vázquez
- Laboratory of Developmental Biology, Instituto Tecnológico de Chascomús (CONICET-UNSAM). Escuela de Bio y Nanotecnologías (UNSAM), Chascomús, Buenos Aires 7130, Argentina
| | - Pablo H Strobl-Mazzulla
- Laboratory of Developmental Biology, Instituto Tecnológico de Chascomús (CONICET-UNSAM). Escuela de Bio y Nanotecnologías (UNSAM), Chascomús, Buenos Aires 7130, Argentina
| |
Collapse
|
27
|
Yao B, Hsu C, Goldner G, Michaeli Y, Ebenstein Y, Listgarten J. Effective training of nanopore callers for epigenetic marks with limited labelled data. Open Biol 2024; 14:230449. [PMID: 38862018 DOI: 10.1098/rsob.230449] [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: 12/12/2023] [Accepted: 03/04/2024] [Indexed: 06/13/2024] Open
Abstract
Nanopore sequencing platforms combined with supervised machine learning (ML) have been effective at detecting base modifications in DNA such as 5-methylcytosine (5mC) and N6-methyladenine (6mA). These ML-based nanopore callers have typically been trained on data that span all modifications on all possible DNA [Formula: see text]-mer backgrounds-a complete training dataset. However, as nanopore technology is pushed to more and more epigenetic modifications, such complete training data will not be feasible to obtain. Nanopore calling has historically been performed with hidden Markov models (HMMs) that cannot make successful calls for [Formula: see text]-mer contexts not seen during training because of their independent emission distributions. However, deep neural networks (DNNs), which share parameters across contexts, are increasingly being used as callers, often outperforming their HMM cousins. It stands to reason that a DNN approach should be able to better generalize to unseen [Formula: see text]-mer contexts. Indeed, herein we demonstrate that a common DNN approach (DeepSignal) outperforms a common HMM approach (Nanopolish) in the incomplete data setting. Furthermore, we propose a novel hybrid HMM-DNN approach, amortized-HMM, that outperforms both the pure HMM and DNN approaches on 5mC calling when the training data are incomplete. This type of approach is expected to be useful for calling other base modifications such as 5-hydroxymethylcytosine and for the simultaneous calling of different modifications, settings in which complete training data are not likely to be available.
Collapse
Affiliation(s)
- Brian Yao
- Department of Electrical Engineering & Computer Sciences, University of California , Berkeley, CA 94720, USA
| | - Chloe Hsu
- Department of Electrical Engineering & Computer Sciences, University of California , Berkeley, CA 94720, USA
| | - Gal Goldner
- Department of Chemical Physics, Tel Aviv University , Tel Aviv-Yafo, Israel
| | - Yael Michaeli
- Department of Chemical Physics, Tel Aviv University , Tel Aviv-Yafo, Israel
| | - Yuval Ebenstein
- Department of Chemical Physics, Tel Aviv University , Tel Aviv-Yafo, Israel
- Edmond J. Safra Center for Bioinformatics, Tel Aviv University , Tel Aviv-Yafo, Israel
| | - Jennifer Listgarten
- Department of Electrical Engineering & Computer Sciences, University of California , Berkeley, CA 94720, USA
- Center for Computational Biology, University of California , Berkeley, CA 94720, USA
| |
Collapse
|
28
|
Andrade-Brito DE, Núñez-Ríos DL, Martínez-Magaña JJ, Nagamatsu ST, Rompala G, Zillich L, Witt SH, Clark SL, Lattig MC, Montalvo-Ortiz JL. Neuronal-specific methylome and hydroxymethylome analysis reveal significant loci associated with alcohol use disorder. Front Genet 2024; 15:1345410. [PMID: 38633406 PMCID: PMC11021708 DOI: 10.3389/fgene.2024.1345410] [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: 11/27/2023] [Accepted: 03/14/2024] [Indexed: 04/19/2024] Open
Abstract
Background: Alcohol use disorder (AUD) is a complex condition associated with adverse health consequences that affect millions of individuals worldwide. Epigenetic modifications, including DNA methylation (5 mC), have been associated with AUD and other alcohol-related traits. Epigenome-wide association studies (EWAS) have identified differentially methylated genes associated with AUD in human peripheral and brain tissue. More recently, epigenetic studies of AUD have also evaluated DNA hydroxymethylation (5 hmC) in the human brain. However, most of the epigenetic work in postmortem brain tissue has examined bulk tissue. In this study, we investigated neuronal-specific 5 mC and 5 hmC alterations at CpG sites associated with AUD in the human orbitofrontal cortex (OFC). Methods: Neuronal nuclei from the OFC were evaluated in 34 human postmortem brain samples (10 AUD, 24 non-AUD). Reduced representation oxidative bisulfite sequencing was used to assess 5 mC and 5 hmC at the genome-wide level. Differential 5 mC and 5 hmC were evaluated using the methylKit R package and significance was set at false discovery rate < 0.05 and differential methylation > 2. Functional enrichment analyses were performed, and gene-level convergence was evaluated in an independent dataset that assessed 5 mC and 5 hmC of AUD in bulk cortical tissue. Results: We identified 417 5 mC and 363 5hmC significant differential CpG sites associated with AUD, with 59% in gene promoters. Some of the identified genes have been previously implicated in alcohol consumption, including SYK, DNMT3A for 5 mC, GAD1, DLX1, DLX2, for 5 hmC and GATA4 in both. Convergence with a previous AUD 5 mC and 5 hmC study was observed for 28 genes. We also identified 5 and 35 differential regions for 5 mC and 5 hmC, respectively. Lastly, GWAS enrichment analysis showed an association with AUD for differential 5 mC genes. Discussion: This study reveals neuronal-specific methylome and hydroxymethylome dysregulation associated with AUD, identifying both previously reported and potentially novel gene associations with AUD. Our findings provide new insights into the epigenomic dysregulation of AUD in the human brain.
Collapse
Affiliation(s)
- Diego E. Andrade-Brito
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
- National Center of Post-Traumatic Stress Disorder, VA CT Healthcare, West Haven, CT, United States
| | - Diana L. Núñez-Ríos
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
- National Center of Post-Traumatic Stress Disorder, VA CT Healthcare, West Haven, CT, United States
| | - José Jaime Martínez-Magaña
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
- National Center of Post-Traumatic Stress Disorder, VA CT Healthcare, West Haven, CT, United States
| | - Sheila T. Nagamatsu
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
- National Center of Post-Traumatic Stress Disorder, VA CT Healthcare, West Haven, CT, United States
| | - Gregory Rompala
- Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| | - Lea Zillich
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Stephanie H. Witt
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Shaunna L. Clark
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, College Station, TX, United States
| | - Maria C. Lattig
- Facultad de Ciencias, Universidad de los Andes, Bogotá, Colombia
| | - Janitza L. Montalvo-Ortiz
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
- National Center of Post-Traumatic Stress Disorder, VA CT Healthcare, West Haven, CT, United States
| |
Collapse
|
29
|
Bordin DL, Grooms K, Montaldo NP, Fordyce Martin SL, Sætrom P, Samson LD, Bjørås M, van Loon B. Loss of alkyladenine DNA glycosylase alters gene expression in the developing mouse brain and leads to reduced anxiety and improved memory. DNA Repair (Amst) 2024; 135:103632. [PMID: 38280242 DOI: 10.1016/j.dnarep.2024.103632] [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: 10/06/2023] [Revised: 01/05/2024] [Accepted: 01/19/2024] [Indexed: 01/29/2024]
Abstract
Neurodevelopment is a tightly coordinated process, during which the genome is exposed to spectra of endogenous agents at different stages of differentiation. Emerging evidence indicates that DNA damage is an important feature of developing brain, tightly linked to gene expression and neuronal activity. Some of the most frequent DNA damage includes changes to DNA bases, which are recognized by DNA glycosylases and repaired through base excision repair (BER) pathway. The only mammalian DNA glycosylase able to remove frequent alkylated DNA based is alkyladenine DNA glycosylase (Aag, aka Mpg). We recently demonstrated that, besides its role in DNA repair, AAG affects expression of neurodevelopmental genes in human cells. Aag was further proposed to act as reader of epigenetic marks, including 5-hydroxymethylcytosine (5hmC), in the mouse brain. Despite the potential Aag involvement in the key brain processes, the impact of Aag loss on developing brain remains unknown. Here, by using Aag knockout (Aag-/-) mice, we show that Aag absence leads to reduced DNA break levels, evident in lowered number of γH2AX foci in postnatal day 5 (P5) hippocampi. This is accompanied by changes in 5hmC signal intensity in different hippocampal regions. Transcriptome analysis of hippocampi and prefrontal cortex, at different developmental stages, indicates that lack of Aag alters gene expression, primarily of genes involved in regulation of response to stress. Across all developmental stages tested aldehyde dehydrogenase 2 (Aldh2) emerged as one of the most prominent genes deregulated in Aag-dependent manner. In line with the changes in hippocampal DNA damage levels and the gene expression, adult Aag-/- mice exhibit altered behavior, evident in decreased anxiety levels determined in the Elevated Zero Maze and increased alternations in the Elevated T Maze tests. Taken together these results suggests that Aag has functions in modulation of genome dynamics during brain development, important for animal behavior.
Collapse
Affiliation(s)
- Diana L Bordin
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, 7049 Trondheim, Norway
| | - Kayla Grooms
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, 7049 Trondheim, Norway
| | - Nicola P Montaldo
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, 7049 Trondheim, Norway; Department of Microbiology, Oslo University Hospital, University of Oslo, Oslo 0372, Norway
| | - Sarah L Fordyce Martin
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, 7049 Trondheim, Norway
| | - Pål Sætrom
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, 7049 Trondheim, Norway; Bioinformatics core facility - BioCore; Norwegian University of Science and Technology (NTNU), 7491 Trondheim, Norway; K.G. Jebsen Center for Genetic Epidemiology, Norwegian University of Science and Technology (NTNU), 7491 Trondheim, Norway; Department of Computer Science, Faculty of Information Technology and Electrical Engineering, Norwegian University of Science and Technology (NTNU), 7491 Trondheim, Norway
| | - Leona D Samson
- Department of Biological Engineering, Department of Biology, David H. Koch Institute of integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Magnar Bjørås
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, 7049 Trondheim, Norway; Department of Microbiology, Oslo University Hospital, University of Oslo, Oslo 0372, Norway
| | - Barbara van Loon
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, 7049 Trondheim, Norway.
| |
Collapse
|
30
|
Sugawara H, Date A, Fuke S, Nakachi Y, Kato T, Narita M, Bundo M, Iwamoto K. Quantification of cytosine modifications in the aged mouse brain. Neuropsychopharmacol Rep 2024; 44:250-255. [PMID: 38058257 PMCID: PMC10932792 DOI: 10.1002/npr2.12396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 10/18/2023] [Accepted: 10/25/2023] [Indexed: 12/08/2023] Open
Abstract
Quantifying cytosine modifications in various brain regions provides important insights into the gene expression regulation and pathophysiology of neuropsychiatric disorders. In this study, we quantified 5-methylcytosine (5-mC), 5-hydroxymethylation (5-hmC), and 5-formylcytosine (5-fC) levels in five brain regions (the frontal lobe, cerebral cortical region without frontal lobe, hippocampus, basal ganglia, and the cerebellum) and the heart at three developmental periods (12, 48, and 101 weeks). We observed significant regional variations in cytosine modification. Notably, regional variations were generally maintained throughout development, suggesting that epigenetic regulation is unique to each brain region and remains relatively stable with age. The 5-mC and 5-hmC levels were positively correlated, although the extent of the correlations seemed to differ in different brain regions. On the contrary, 5-fC levels did not correlate with 5-mC or 5-hmC levels. Additionally, we observed an age-dependent decrease in 5-fC levels in the basal ganglia, suggesting a unique epigenetic regulation mechanism. Further high-resolution studies using animal models of neuropsychiatric disorders as well as postmortem brain evaluation are warranted.
Collapse
Affiliation(s)
- Hiroko Sugawara
- Department of Psychiatry, Faculty of MedicineFukuoka UniversityFukuokaJapan
- Department of PsychiatryKansai Rosai HospitalAmagasakiJapan
- Department of Psychiatry, Graduate School of MedicineOsaka UniversityOsakaJapan
| | - Akitoshi Date
- Department of PharmacologyHoshi University School of Pharmacy and Pharmaceutical SciencesTokyoJapan
| | - Satoshi Fuke
- Lab for Molecular Dynamics of Mental DisordersRIKEN Center for Brain ScienceWakoJapan
- Research Unit/NeuroscienceSohyaku. Innovative Research Division, Mitsubishi Tanabe Pharma CorporationYokohamaJapan
| | - Yutaka Nakachi
- Department of Molecular Brain Science, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Tadafumi Kato
- Lab for Molecular Dynamics of Mental DisordersRIKEN Center for Brain ScienceWakoJapan
- Department of Psychiatry and Behavior Science, Graduate School of MedicineJuntendo UniversityTokyoJapan
| | - Minoru Narita
- Department of PharmacologyHoshi University School of Pharmacy and Pharmaceutical SciencesTokyoJapan
| | - Miki Bundo
- Lab for Molecular Dynamics of Mental DisordersRIKEN Center for Brain ScienceWakoJapan
- Department of Molecular Brain Science, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Kazuya Iwamoto
- Lab for Molecular Dynamics of Mental DisordersRIKEN Center for Brain ScienceWakoJapan
- Department of Molecular Brain Science, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| |
Collapse
|
31
|
Bijlani S, Pang KM, Bugga LV, Rangasamy S, Narayanan V, Chatterjee S. Nuclease-free precise genome editing corrects MECP2 mutations associated with Rett syndrome. Front Genome Ed 2024; 6:1346781. [PMID: 38495533 PMCID: PMC10940404 DOI: 10.3389/fgeed.2024.1346781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 02/12/2024] [Indexed: 03/19/2024] Open
Abstract
Rett syndrome is an acquired progressive neurodevelopmental disorder caused by de novo mutations in the X-linked MECP2 gene which encodes a pleiotropic protein that functions as a global transcriptional regulator and a chromatin modifier. Rett syndrome predominantly affects heterozygous females while affected male hemizygotes rarely survive. Gene therapy of Rett syndrome has proven challenging due to a requirement for stringent regulation of expression with either over- or under-expression being toxic. Ectopic expression of MECP2 in conjunction with regulatory miRNA target sequences has achieved some success, but the durability of this approach remains unknown. Here we evaluated a nuclease-free homologous recombination (HR)-based genome editing strategy to correct mutations in the MECP2 gene. The stem cell-derived AAVHSCs have previously been shown to mediate seamless and precise HR-based genome editing. We tested the ability of HR-based genome editing to correct pathogenic mutations in Exons 3 and 4 of the MECP2 gene and restore the wild type sequence while preserving all native genomic regulatory elements associated with MECP2 expression, thus potentially addressing a significant issue in gene therapy for Rett syndrome. Moreover, since the mutations are edited directly at the level of the genome, the corrections are expected to be durable with progeny cells inheriting the edited gene. The AAVHSC MECP2 editing vector was designed to be fully homologous to the target MECP2 region and to insert a promoterless Venus reporter at the end of Exon 4. Evaluation of AAVHSC editing in a panel of Rett cell lines bearing mutations in Exons 3 and 4 demonstrated successful correction and rescue of expression of the edited MECP2 gene. Sequence analysis of edited Rett cells revealed successful and accurate correction of mutations in both Exons 3 and 4 and permitted mapping of HR crossover events. Successful correction was observed only when the mutations were flanked at both the 5' and 3' ends by crossover events, but not when both crossovers occurred either exclusively upstream or downstream of the mutation. Importantly, we concluded that pathogenic mutations were successfully corrected in every Rett line analyzed, demonstrating the therapeutic potential of HR-based genome editing.
Collapse
Affiliation(s)
- Swati Bijlani
- Department of Surgery, Beckman Research Institute of the City of Hope, Duarte, CA, United States
| | - Ka Ming Pang
- Department of Surgery, Beckman Research Institute of the City of Hope, Duarte, CA, United States
| | - Lakshmi V. Bugga
- Department of Surgery, Beckman Research Institute of the City of Hope, Duarte, CA, United States
| | - Sampath Rangasamy
- Center for Rare Childhood Disorders (C4RCD), Neurogenomics Division, Translational Genomics Research Institute (TGen), Phoenix, AZ, United States
| | - Vinodh Narayanan
- Center for Rare Childhood Disorders (C4RCD), Neurogenomics Division, Translational Genomics Research Institute (TGen), Phoenix, AZ, United States
| | - Saswati Chatterjee
- Department of Surgery, Beckman Research Institute of the City of Hope, Duarte, CA, United States
| |
Collapse
|
32
|
Martinez D, Jiang E, Zhou Z. Overcoming genetic and cellular complexity to study the pathophysiology of X-linked intellectual disabilities. J Neurodev Disord 2024; 16:5. [PMID: 38424476 PMCID: PMC10902969 DOI: 10.1186/s11689-024-09517-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 02/04/2024] [Indexed: 03/02/2024] Open
Abstract
X-linked genetic causes of intellectual disability (ID) account for a substantial proportion of cases and remain poorly understood, in part due to the heterogeneous expression of X-linked genes in females. This is because most genes on the X chromosome are subject to random X chromosome inactivation (XCI) during early embryonic development, which results in a mosaic pattern of gene expression for a given X-linked mutant allele. This mosaic expression produces substantial complexity, especially when attempting to study the already complicated neural circuits that underly behavior, thus impeding the understanding of disease-related pathophysiology and the development of therapeutics. Here, we review a few selected X-linked forms of ID that predominantly affect heterozygous females and the current obstacles for developing effective therapies for such disorders. We also propose a genetic strategy to overcome the complexity presented by mosaicism in heterozygous females and highlight specific tools for studying synaptic and circuit mechanisms, many of which could be shared across multiple forms of intellectual disability.
Collapse
Affiliation(s)
- Dayne Martinez
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19102, USA
- Medical Scientist Training Program, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19102, USA
| | - Evan Jiang
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19102, USA
- Medical Scientist Training Program, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19102, USA
| | - Zhaolan Zhou
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19102, USA.
- Medical Scientist Training Program, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19102, USA.
- Department of Neuroscience, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19102, USA.
- Intellectual and Developmental Disabilities Research Center, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA.
| |
Collapse
|
33
|
Friese S, Ranzini G, Tuchtenhagen M, Lossow K, Hertel B, Pohl G, Ebert F, Bornhorst J, Kipp AP, Schwerdtle T. Long-term suboptimal dietary trace element supply does not affect trace element homeostasis in murine cerebellum. Metallomics 2024; 16:mfae003. [PMID: 38299785 PMCID: PMC10873500 DOI: 10.1093/mtomcs/mfae003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 12/14/2023] [Indexed: 02/02/2024]
Abstract
The ageing process is associated with alterations of systemic trace element (TE) homeostasis increasing the risk, e.g. neurodegenerative diseases. Here, the impact of long-term modulation of dietary intake of copper, iron, selenium, and zinc was investigated in murine cerebellum. Four- and 40-wk-old mice of both sexes were supplied with different amounts of those TEs for 26 wk. In an adequate supply group, TE concentrations were in accordance with recommendations for laboratory mice while suboptimally supplied animals received only limited amounts of copper, iron, selenium, and zinc. An additional age-adjusted group was fed selenium and zinc in amounts exceeding recommendations. Cerebellar TE concentrations were measured by inductively coupled plasma-tandem mass spectrometry. Furthermore, the expression of genes involved in TE transport, DNA damage response, and DNA repair as well as selected markers of genomic stability [8-oxoguanine, incision efficiency toward 8-oxoguanine, 5-hydroxyuracil, and apurinic/apyrimidinic sites and global DNA (hydroxy)methylation] were analysed. Ageing resulted in a mild increase of iron and copper concentrations in the cerebellum, which was most pronounced in the suboptimally supplied groups. Thus, TE changes in the cerebellum were predominantly driven by age and less by nutritional intervention. Interestingly, deviation from adequate TE supply resulted in higher manganese concentrations of female mice even though the manganese supply itself was not modulated. Parameters of genomic stability were neither affected by age, sex, nor diet. Overall, this study revealed that suboptimal dietary TE supply does not substantially affect TE homeostasis in the murine cerebellum.
Collapse
Affiliation(s)
- Sharleen Friese
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
- TraceAge—DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena-Wuppertal, Germany
| | - Giovanna Ranzini
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
| | - Max Tuchtenhagen
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
- TraceAge—DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena-Wuppertal, Germany
| | - Kristina Lossow
- TraceAge—DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena-Wuppertal, Germany
- Nutritional Physiology, Institute of Nutritional Sciences, Friedrich Schiller University Jena, Dornburger Str. 24, 07743 Jena, Germany
| | - Barbara Hertel
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
| | - Gabriele Pohl
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
| | - Franziska Ebert
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
| | - Julia Bornhorst
- TraceAge—DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena-Wuppertal, Germany
- Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Gaußstraße 20, 42119 Wuppertal, Germany
| | - Anna Patricia Kipp
- TraceAge—DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena-Wuppertal, Germany
- Nutritional Physiology, Institute of Nutritional Sciences, Friedrich Schiller University Jena, Dornburger Str. 24, 07743 Jena, Germany
| | - Tanja Schwerdtle
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
- TraceAge—DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena-Wuppertal, Germany
- German Federal Institute for Risk Assessment (BfR), Max-Dohrn-Str. 8-10, 10589 Berlin, Germany
| |
Collapse
|
34
|
Ehrlich M, Ehrlich KC, Lacey M, Baribault C, Sen S, Estève PO, Pradhan S. Epigenetics of Genes Preferentially Expressed in Dissimilar Cell Populations: Myoblasts and Cerebellum. EPIGENOMES 2024; 8:4. [PMID: 38390894 PMCID: PMC10885033 DOI: 10.3390/epigenomes8010004] [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: 12/12/2023] [Revised: 01/23/2024] [Accepted: 01/23/2024] [Indexed: 02/24/2024] Open
Abstract
While studying myoblast methylomes and transcriptomes, we found that CDH15 had a remarkable preference for expression in both myoblasts and cerebellum. To understand how widespread such a relationship was and its epigenetic and biological correlates, we systematically looked for genes with similar transcription profiles and analyzed their DNA methylation and chromatin state and accessibility profiles in many different cell populations. Twenty genes were expressed preferentially in myoblasts and cerebellum (Myob/Cbl genes). Some shared DNA hypo- or hypermethylated regions in myoblasts and cerebellum. Particularly striking was ZNF556, whose promoter is hypomethylated in expressing cells but highly methylated in the many cell populations that do not express the gene. In reporter gene assays, we demonstrated that its promoter's activity is methylation sensitive. The atypical epigenetics of ZNF556 may have originated from its promoter's hypomethylation and selective activation in sperm progenitors and oocytes. Five of the Myob/Cbl genes (KCNJ12, ST8SIA5, ZIC1, VAX2, and EN2) have much higher RNA levels in cerebellum than in myoblasts and displayed myoblast-specific hypermethylation upstream and/or downstream of their promoters that may downmodulate expression. Differential DNA methylation was associated with alternative promoter usage for Myob/Cbl genes MCF2L, DOK7, CNPY1, and ANK1. Myob/Cbl genes PAX3, LBX1, ZNF556, ZIC1, EN2, and VAX2 encode sequence-specific transcription factors, which likely help drive the myoblast and cerebellum specificity of other Myob/Cbl genes. This study extends our understanding of epigenetic/transcription associations related to differentiation and may help elucidate relationships between epigenetic signatures and muscular dystrophies or cerebellar-linked neuropathologies.
Collapse
Affiliation(s)
- Melanie Ehrlich
- Tulane Cancer Center, Hayward Human Genetics Center, Center for Bioinformatics and Genomics, Tulane University Health Sciences Center, New Orleans, LA 70112, USA
- Center for Bioinformatics and Genomics, Tulane University Health Sciences Center, New Orleans, LA 70112, USA
| | - Kenneth C Ehrlich
- Center for Bioinformatics and Genomics, Tulane University Health Sciences Center, New Orleans, LA 70112, USA
| | - Michelle Lacey
- Department of Mathematics, Tulane University, New Orleans, LA 70118, USA
| | - Carl Baribault
- Information Technology, Tulane University, New Orleans, LA 70118, USA
| | - Sagnik Sen
- Genome Biology Division, New England Biolabs, Ipswich, MA 01938, USA
| | | | - Sriharsa Pradhan
- Genome Biology Division, New England Biolabs, Ipswich, MA 01938, USA
| |
Collapse
|
35
|
Xie J, Wang Y, Ye C, Li XJ, Lin L. Distinctive Patterns of 5-Methylcytosine and 5-Hydroxymethylcytosine in Schizophrenia. Int J Mol Sci 2024; 25:636. [PMID: 38203806 PMCID: PMC10779130 DOI: 10.3390/ijms25010636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 12/25/2023] [Accepted: 12/30/2023] [Indexed: 01/12/2024] Open
Abstract
Schizophrenia is a highly heritable neuropsychiatric disorder characterized by cognitive and social dysfunction. Genetic, epigenetic, and environmental factors are together implicated in the pathogenesis and development of schizophrenia. DNA methylation, 5-methycytosine (5mC) and 5-hydroxylcytosine (5hmC) have been recognized as key epigenetic elements in neurodevelopment, ageing, and neurodegenerative diseases. Recently, distinctive 5mC and 5hmC pattern and expression changes of related genes have been discovered in schizophrenia. Antipsychotic drugs that affect 5mC status can alleviate symptoms in patients with schizophrenia, suggesting a critical role for DNA methylation in the pathogenesis of schizophrenia. Further exploring the signatures of 5mC and 5hmC in schizophrenia and developing precision-targeted epigenetic drugs based on this will provide new insights into the diagnosis and treatment of schizophrenia.
Collapse
Affiliation(s)
| | | | | | | | - Li Lin
- Guangdong Key Laboratory of Non-Human Primate Research, Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China; (J.X.); (Y.W.); (C.Y.); (X.-J.L.)
| |
Collapse
|
36
|
Wang Z, Ma H, Nasir A, Liu S, Li Z, Tao F, Bai Q. TET1-mediated epigenetic regulation of tumor necrosis factor-α in trigeminal ganglia contributes to chronic temporomandibular joint pain. Life Sci 2024; 336:122283. [PMID: 37993094 DOI: 10.1016/j.lfs.2023.122283] [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: 10/03/2023] [Revised: 11/15/2023] [Accepted: 11/19/2023] [Indexed: 11/24/2023]
Abstract
Chronic temporomandibular joint (TMJ) pain profoundly affects patients' quality of life. Trigeminal tumor necrosis factor-α (TNFα) plays a pivotal role in mediating TMJ pain in mice, yet the underlying epigenetic mechanisms remain enigmatic. To unravel these epigenetic intricacies, we employed a multifaceted approach. Hydroxymethylated DNA immunoprecipitation (hMeDIP) and chromatin immunoprecipitation (ChIP) followed by qPCR were employed to investigate the demethylation of TNFα gene (Tnfa) and its regulation by ten-eleven translocation methylcytosine dioxygenase 1 (TET1) in a chronic TMJ pain mouse model. The global levels of 5-hydroxymethylcytosine (5hmc) and percentage of 5hmc at the Tnfa promoter region were measured in the trigeminal ganglia (TG) and spinal trigeminal nucleus caudalis (Sp5C) following complete Freund's adjuvant (CFA) or saline treatment. TET1 knockdown and pain behavioral testing were conducted to ascertain the role of TET1-mediated epigenetic regulation of TNFα in the pathogenesis of chronic TMJ pain. Our finding revealed an increase in 5hmc at the Tnfa promoter region in both TG and Sp5C of CFA-treated mice. TET1 was upregulated in the mouse TG, and the ChIP result showed TET1 direct binding to the Tnfa promoter, with higher efficiency in the CFA-treated group. Immunofluorescence revealed the predominant expression of TET1 in trigeminal neurons. TET1 knockdown in the TG significantly reversed CFA-induced TNFα upregulation and alleviated chronic TMJ pain. In conclusion, our study implicates TET1 as a vital epigenetic regulator contributing to chronic inflammatory TMJ pain via trigeminal TNFα signaling. Targeting TET1 holds promise for epigenetic interventions in TMJ pain management.
Collapse
Affiliation(s)
- Zhitao Wang
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Heng Ma
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Abdul Nasir
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China; Medical Research Center, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Sufang Liu
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX, USA
| | - Zhisong Li
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Feng Tao
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX, USA.
| | - Qian Bai
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China; Medical Research Center, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
37
|
Ma Y, Wang W, Liu S, Qiao X, Xing Y, Zhou Q, Zhang Z. Epigenetic Regulation of Neuroinflammation in Alzheimer's Disease. Cells 2023; 13:79. [PMID: 38201283 PMCID: PMC10778497 DOI: 10.3390/cells13010079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 12/25/2023] [Accepted: 12/28/2023] [Indexed: 01/12/2024] Open
Abstract
Alzheimer's disease (AD) is a chronic and progressive neurodegenerative disease and clinically manifests with cognitive decline and behavioral disabilities. Over the past years, mounting studies have demonstrated that the inflammatory response plays a key role in the onset and development of AD, and neuroinflammation has been proposed as the third major pathological driving factor of AD, ranking after the two well-known core pathologies, amyloid β (Aβ) deposits and neurofibrillary tangles (NFTs). Epigenetic mechanisms, referring to heritable changes in gene expression independent of DNA sequence alterations, are crucial regulators of neuroinflammation which have emerged as potential therapeutic targets for AD. Upon regulation of transcriptional repression or activation, epigenetic modification profiles are closely involved in inflammatory gene expression and signaling pathways of neuronal differentiation and cognitive function in central nervous system disorders. In this review, we summarize the current knowledge about epigenetic control mechanisms with a focus on DNA and histone modifications involved in the regulation of inflammatory genes and signaling pathways in AD, and the inhibitors under clinical assessment are also discussed.
Collapse
Affiliation(s)
- Yajing Ma
- College of Biology and Food, Shangqiu Normal University, Shangqiu 476000, China;
| | - Wang Wang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (W.W.); (Y.X.)
| | - Sufang Liu
- Department of Biomedical Sciences, College of Dentistry, Texas A&M University, Dallas, TX 75246, USA;
| | - Xiaomeng Qiao
- Department of Pathology and Forensic Medicine, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China;
| | - Ying Xing
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (W.W.); (Y.X.)
| | - Qingfeng Zhou
- College of Biology and Food, Shangqiu Normal University, Shangqiu 476000, China;
| | - Zhijian Zhang
- College of Biology and Food, Shangqiu Normal University, Shangqiu 476000, China;
| |
Collapse
|
38
|
Yu D, Li M, Linghu G, Hu Y, Hajdarovic KH, Wang A, Singh R, Webb AE. CellBiAge: Improved single-cell age classification using data binarization. Cell Rep 2023; 42:113500. [PMID: 38032797 PMCID: PMC10791072 DOI: 10.1016/j.celrep.2023.113500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 10/20/2023] [Accepted: 11/13/2023] [Indexed: 12/02/2023] Open
Abstract
Aging is a major risk factor for many diseases. Accurate methods for predicting age in specific cell types are essential to understand the heterogeneity of aging and to assess rejuvenation strategies. However, classifying organismal age at single-cell resolution using transcriptomics is challenging due to sparsity and noise. Here, we developed CellBiAge, a robust and easy-to-implement machine learning pipeline, to classify the age of single cells in the mouse brain using single-cell transcriptomics. We show that binarization of gene expression values for the top highly variable genes significantly improved test performance across different models, techniques, sexes, and brain regions, with potential age-related genes identified for model prediction. Additionally, we demonstrate CellBiAge's ability to capture exercise-induced rejuvenation in neural stem cells. This study provides a broadly applicable approach for robust classification of organismal age of single cells in the mouse brain, which may aid in understanding the aging process and evaluating rejuvenation methods.
Collapse
Affiliation(s)
- Doudou Yu
- Molecular Biology, Cell Biology, and Biochemistry Graduate Program, Brown University, Providence, RI 02912, USA; Data Science Institute, Brown University, Providence, RI 02912, USA
| | - Manlin Li
- Data Science Institute, Brown University, Providence, RI 02912, USA
| | - Guanjie Linghu
- Data Science Institute, Brown University, Providence, RI 02912, USA
| | - Yihuan Hu
- Data Science Institute, Brown University, Providence, RI 02912, USA
| | | | - An Wang
- Department of Applied Mathematics & Statistics, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Ritambhara Singh
- Department of Computer Science, Brown University, Providence, RI 02912, USA; Center for Computational Molecular Biology, Brown University, Providence, RI 02912, USA.
| | - Ashley E Webb
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI 02912, USA; Center on the Biology of Aging, Brown University, Providence, RI 02912, USA; Carney Institute for Brain Science, Brown University, Providence, RI 02912, USA; Center for Translational Neuroscience, Brown University, Providence, RI 02912, USA.
| |
Collapse
|
39
|
Zhuo L, Wang R, Fu X, Yao X. StableDNAm: towards a stable and efficient model for predicting DNA methylation based on adaptive feature correction learning. BMC Genomics 2023; 24:742. [PMID: 38053026 DOI: 10.1186/s12864-023-09802-7] [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: 07/26/2023] [Accepted: 11/11/2023] [Indexed: 12/07/2023] Open
Abstract
BACKGROUND DNA methylation, instrumental in numerous life processes, underscores the paramount importance of its accurate prediction. Recent studies suggest that deep learning, due to its capacity to extract profound insights, provides a more precise DNA methylation prediction. However, issues related to the stability and generalization performance of these models persist. RESULTS In this study, we introduce an efficient and stable DNA methylation prediction model. This model incorporates a feature fusion approach, adaptive feature correction technology, and a contrastive learning strategy. The proposed model presents several advantages. First, DNA sequences are encoded at four levels to comprehensively capture intricate information across multi-scale and low-span features. Second, we design a sequence-specific feature correction module that adaptively adjusts the weights of sequence features. This improvement enhances the model's stability and scalability, or its generality. Third, our contrastive learning strategy mitigates the instability issues resulting from sparse data. To validate our model, we conducted multiple sets of experiments on commonly used datasets, demonstrating the model's robustness and stability. Simultaneously, we amalgamate various datasets into a single, unified dataset. The experimental outcomes from this combined dataset substantiate the model's robust adaptability. CONCLUSIONS Our research findings affirm that the StableDNAm model is a general, stable, and effective instrument for DNA methylation prediction. It holds substantial promise for providing invaluable assistance in future methylation-related research and analyses.
Collapse
Affiliation(s)
- Linlin Zhuo
- College of Data Science and Artificial Intelligence, Wenzhou University of Technology, Wenzhou, 325000, China
| | - Rui Wang
- College of Data Science and Artificial Intelligence, Wenzhou University of Technology, Wenzhou, 325000, China
| | - Xiangzheng Fu
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, 410000, China.
| | - Xiaojun Yao
- Faculty of Applied Sciences, Macao Polytechnic University, Macao, 999078, China.
| |
Collapse
|
40
|
Andrade-Brito DE, Núñez-Ríos DL, Martínez-Magaña JJ, Nagamatsu ST, Rompala G, Zillich L, Witt SH, Clark SL, Latig MC, Traumatic Stress Brain Research Group, PGC SUD Epigenetics Working Group, Montalvo-Ortiz JL. Neuronal-specific methylome and hydroxymethylome analysis reveal replicated and novel loci associated with alcohol use disorder. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.11.28.23299094. [PMID: 38105948 PMCID: PMC10725575 DOI: 10.1101/2023.11.28.23299094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Alcohol use disorder (AUD) is a complex condition associated with adverse health consequences that affect millions of individuals worldwide. Epigenetic modifications, including DNA methylation (5mC), have been associated with AUD and other alcohol-related traits. Epigenome-wide association studies (EWAS) have identified differentially methylated genes associated with AUD in human peripheral and brain tissue. More recently, epigenetic studies of AUD have also evaluated DNA hydroxymethylation (5hmC) in the human brain. However, most of the epigenetic work in postmortem brain tissue has examined bulk tissue. In this study, we investigated neuronal-specific 5mC and 5hmC alterations at CpG sites associated with AUD in the human orbitofrontal cortex (OFC). Neuronal nuclei from the OFC were evaluated in 34 human postmortem brain samples (10 AUD, 24 non-AUD). Reduced representation oxidative bisulfite sequencing was used to assess 5mC and 5hmC at the genome-wide level. Differential 5mC and 5hmC were evaluated using the methylKit R package and significance was set at false discovery rate <0.05 and differential methylation >2. Functional enrichment analyses were performed and replication was evaluated replication in an independent dataset that assessed 5mC and 5hmC of AUD in bulk cortical tissue. We identified 417 5mC and 363 5hmC genome-wide significant differential CpG sites associated with AUD, with 59% in gene promoters. We also identified genes previously implicated in alcohol consumption, such as SYK, CHRM2, DNMT3A, and GATA4, for 5mC and GATA4, and GAD1, GATA4, DLX1 for 5hmC. Replication was observed for 28 CpG sites from a previous AUD 5mC and 5hmC study, including FOXP1. Lastly, GWAS enrichment analysis showed an association with AUD for differential 5mC genes. This study reveals neuronal-specific methylome and hydroxymethylome dysregulation associated with AUD. We replicated previous findings and identified novel associations with AUD for both 5mC and 5hmC marks within the OFC. Our findings provide new insights into the epigenomic dysregulation of AUD in the human brain.
Collapse
Affiliation(s)
- Diego E. Andrade-Brito
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- National Center of Post-Traumatic Stress Disorder, VA CT Healthcare, West Haven, CT, USA
| | - Diana L. Núñez-Ríos
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- National Center of Post-Traumatic Stress Disorder, VA CT Healthcare, West Haven, CT, USA
| | - José Jaime Martínez-Magaña
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- National Center of Post-Traumatic Stress Disorder, VA CT Healthcare, West Haven, CT, USA
| | - Sheila T. Nagamatsu
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- National Center of Post-Traumatic Stress Disorder, VA CT Healthcare, West Haven, CT, USA
| | - Gregory Rompala
- Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Lea Zillich
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Stephanie H. Witt
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Shaunna L. Clark
- Department of Psychiatry & Behavioral Sciences, Texas A&M University, College Station, Texas, USA
| | - Maria C. Latig
- Facultad de Ciencias, Universidad de los Andes, Bogotá, Colombia
| | | | - Janitza L. Montalvo-Ortiz
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- National Center of Post-Traumatic Stress Disorder, VA CT Healthcare, West Haven, CT, USA
| |
Collapse
|
41
|
Wu D, Li Y, Li C, Zhong S, Liu B, Hang H, Wang H. MDM2 Antagonist Nutlin-3 Stimulates Global DNA Hydroxymethylation by Enhancing p53-TET1 Signaling Axis. ACS Chem Biol 2023; 18:2240-2248. [PMID: 37463352 DOI: 10.1021/acschembio.3c00247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
DNA hydroxymethylation is involved in many biological processes, including nuclear reprogramming, embryonic development, and tumor suppression. In this study, we report that an anticancer agent, nutlin-3, selectively stimulates global DNA hydroxymethylation in TP53 wild-type cancer cells as manifested by the elevation of 5-hydroxymethylcytosine (5hmC) in genomic DNA. In contrast, nutlin 3 fails to enhance DNA hydroxymethylation in TP53-mutated cancer cells. Consistently, nutlin-3 as a MDM2 antagonist only activates wild-type but not mutated TP53. Furthermore, nutlin-3 does not alter the expression of TET1 but slightly reduces the expression of TET2 and TET3 proteins. These TET family proteins are responsible for converting 5-methylcytosine (5mC) to 5hmC. Interestingly, TET1 knockdown could significantly block the nutlin-3-induced DNA hydroxymethylation as well as TP53 and P21 activation. Immunoprecipitation analysis supports that p53 strongly interacts with TET1 proteins. These results suggest that nutlin-3 activates TP53 and promotes p53-TET1 interaction. As positive feedback, the p53-TET1 interaction further enhances p53 activation and promotes apoptosis. Collectively, we demonstrate that nutlin-3 stimulates DNA hydroxymethylation and apoptosis via a positive feedback mechanism.
Collapse
Affiliation(s)
- Danni Wu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- China General Microbiological Culture Collection Center, State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yao Li
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Cuiping Li
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Shangwei Zhong
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Baodong Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Haiying Hang
- Key Laboratory for Protein and Peptide Pharmaceuticals, National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Hailin Wang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- China General Microbiological Culture Collection Center, State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- School of Environment, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310000, P. R. China
| |
Collapse
|
42
|
McFadden T, Carucci I, Farrell K, Fletchall E, Jarome TJ. Hypothalamic DNA 5-hydroxymethylation levels are altered by diet-induced weight gain during the development of obesity in a sex-specific manner. Brain Res 2023; 1817:148478. [PMID: 37422205 PMCID: PMC10529936 DOI: 10.1016/j.brainres.2023.148478] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 06/30/2023] [Accepted: 07/05/2023] [Indexed: 07/10/2023]
Abstract
Obesity is a major health concern that is associated with altered gene transcription in the hypothalamus. However, the mechanisms controlling this gene expression dysregulation remain largely unknown. DNA 5-hydroxymethylation (5-hmC) is a potent transcriptional activator that is expressed at 10 times higher levels in the brain than the periphery. Despite this, no study has examined if DNA 5-hmC is altered in the brain following exposure to obesogenic diets or contributes to abnormal weight gain over time. Here, we used a rodent diet-induced obesity model in combination with quantitative molecular assays and CRISPR-dCas9 manipulations to test the role of hypothalamic DNA 5-hmC in abnormal weight gain in male and female rats. We found that males, but not females, have decreased levels of DNA 5-hmC in the hypothalamus following exposure to a high fat diet, which directly correlate with increased body weight. Short-term exposure to a high fat diet, which does not result in significant weight gain, resulted in decreased hypothalamic DNA 5-hmC levels, suggesting these changes occur prior to obesity development. Moreover, decreases in DNA 5-hmC persist even after the high fat diet is removed, though the extent of this is diet-dependent. Importantly, CRISPR-dCas9-mediated upregulation of DNA 5-hmC enzymes in the male, but not female, ventromedial nucleus of the hypothalamus significantly reduced the percentage of weight gained on the high fat diet relative to controls. These results suggest that hypothalamic DNA 5-hmC is an important sex-specific regulator of abnormal weight gain following exposure to high fat diets.
Collapse
Affiliation(s)
| | - Isabella Carucci
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | | | | | - Timothy J Jarome
- School of Animal Sciences, USA; School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA.
| |
Collapse
|
43
|
Bahabry R, Hauser RM, Sánchez RG, Jago SS, Ianov L, Stuckey RJ, Parrish RR, Hoef LV, Lubin FD. Alterations in DNA 5-hydroxymethylation Patterns in the Hippocampus of an Experimental Model of Refractory Epilepsy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.03.560698. [PMID: 37873276 PMCID: PMC10592907 DOI: 10.1101/2023.10.03.560698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Temporal lobe epilepsy (TLE) is a type of focal epilepsy characterized by spontaneous recurrent seizures originating from the hippocampus. The epigenetic reprogramming hypothesis of epileptogenesis suggests that the development of TLE is associated with alterations in gene transcription changes resulting in a hyperexcitable network in TLE. DNA 5-methylcytosine (5-mC) is an epigenetic mechanism that has been associated with chronic epilepsy. However, the contribution of 5-hydroxymethylcytosine (5-hmC), a product of 5-mC demethylation by the Ten-Eleven Translocation (TET) family proteins in chronic TLE is poorly understood. 5-hmC is abundant in the brain and acts as a stable epigenetic mark altering gene expression through several mechanisms. Here, we found that the levels of bulk DNA 5-hmC but not 5-mC were significantly reduced in the hippocampus of human TLE patients and in the kainic acid (KA) TLE rat model. Using 5-hmC hMeDIP-sequencing, we characterized 5-hmC distribution across the genome and found bidirectional regulation of 5-hmC at intergenic regions within gene bodies. We found that hypohydroxymethylated 5-hmC intergenic regions were associated with several epilepsy-related genes, including Gal , SV2, and Kcnj11 and hyperdroxymethylation 5-hmC intergenic regions were associated with Gad65 , TLR4 , and Bdnf gene expression. Mechanistically, Tet1 knockdown in the hippocampus was sufficient to decrease 5-hmC levels and increase seizure susceptibility following KA administration. In contrast, Tet1 overexpression in the hippocampus resulted in increased 5-hmC levels associated with improved seizure resiliency in response to KA. These findings suggest an important role for 5-hmC as an epigenetic regulator of epilepsy that can be manipulated to influence seizure outcomes.
Collapse
|
44
|
Chu B, Liu Z, Liu Y, Jiang H. The Role of Advanced Parental Age in Reproductive Genetics. Reprod Sci 2023; 30:2907-2919. [PMID: 37171772 PMCID: PMC10556127 DOI: 10.1007/s43032-023-01256-2] [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: 12/13/2022] [Accepted: 04/29/2023] [Indexed: 05/13/2023]
Abstract
The increase of parental reproductive age is a worldwide trend in modern society in recent decades. In general, older parents have a significant impact on reproductive genetics and the health of offspring. In particular, advanced parental age contributes to the increase in the risk of adverse neurodevelopmental outcomes in offspring. However, it is currently under debate how and to what extent the health of future generations was affected by the parental age. In this review, we aimed to (i) provide an overview of the effects of age on the fertility and biology of the reproductive organs of the parents, (ii) highlight the candidate biological mechanisms underlying reproductive genetic alterations, and (iii) discuss the relevance of the effect of parental age on offspring between animal experiment and clinical observation. In addition, we think that the impact of environmental factors on cognitive and emotional development of older offspring will be an interesting direction.
Collapse
Affiliation(s)
- Boling Chu
- Department of Biobank, Suining Central Hospital, Suining, 629000, China
| | - Zhi Liu
- Department of Pathology, Suining Central Hospital, Suining, 629000, China
| | - Yihong Liu
- College of Humanities And Management, Guizhou University of Traditional Chinese Medicine, Guizhou, 550025, China
| | - Hui Jiang
- Department of Biobank, Suining Central Hospital, Suining, 629000, China.
| |
Collapse
|
45
|
Watanabe R, Nakachi Y, Matsubara H, Ueda J, Ishii T, Ukai W, Hashimoto E, Kasai K, Simizu S, Kato T, Bundo M, Iwamoto K. Identification of epigenetically active L1 promoters in the human brain and their relationship with psychiatric disorders. Neurosci Res 2023; 195:37-51. [PMID: 37141946 DOI: 10.1016/j.neures.2023.05.001] [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: 02/02/2023] [Revised: 04/09/2023] [Accepted: 05/01/2023] [Indexed: 05/06/2023]
Abstract
Long interspersed nuclear element-1 (LINE-1, L1) affects the transcriptome landscape in multiple ways. Promoter activity within its 5'UTR plays a critical role in regulating diverse L1 activities. However, the epigenetic status of L1 promoters in adult brain cells and their relationship with psychiatric disorders remain poorly understood. Here, we examined DNA methylation and hydroxymethylation of the full-length L1s in neurons and nonneurons and identified "epigenetically active" L1s. Notably, some of epigenetically active L1s were retrotransposition competent, which even had chimeric transcripts from the antisense promoters at their 5'UTRs. We also identified differentially methylated L1s in the prefrontal cortices of patients with psychiatric disorders. In nonneurons of bipolar disorder patients, one L1 was significantly hypomethylated and showed an inverse correlation with the expression level of the overlapping gene NREP. Finally, we observed that altered DNA methylation levels of L1 in patients with psychiatric disorders were not affected by the surrounding genomic regions but originated from the L1 sequences. These results suggested that altered epigenetic regulation of the L1 5'UTR in the brain was involved in the pathophysiology of psychiatric disorders.
Collapse
Affiliation(s)
- Risa Watanabe
- Department of Molecular Brain Science, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yutaka Nakachi
- Department of Molecular Brain Science, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hikari Matsubara
- Department of Molecular Brain Science, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Junko Ueda
- Department of Molecular Brain Science, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Takao Ishii
- Department of Occupational Therapy, Sapporo Medical University School of Health Sciences, Sapporo, Japan
| | - Wataru Ukai
- Department of Neuropsychiatry, Sapporo Medical University, School of Medicine, Sapporo, Japan
| | - Eri Hashimoto
- Department of Neuropsychiatry, Sapporo Medical University, School of Medicine, Sapporo, Japan
| | - Kiyoto Kasai
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan; The International Research Center for Neurointelligence (WPI-IRCN), University of Tokyo Institutes for Advanced Study (UTIAS), The University of Tokyo, Tokyo, Japan; University of Tokyo Institute for Diversity and Adaptation of Human Mind (UTIDAHM), The University of Tokyo, Tokyo, Japan; UTokyo Center for Integrative Science of Human Behaviour (CiSHuB), The University of Tokyo, Tokyo, Japan
| | - Siro Simizu
- Department of Applied Chemistry, Faculty of Science and Technology, Keio University, Yokohama, Japan
| | - Tadafumi Kato
- Department of Psychiatry and Behavioral Science, Juntendo University Graduate School of Medicine, Tokyo, Japan; Laboratory for Molecular Dynamics of Mental Disorders, RIKEN Brain Science Institute, Wako, Saitama, Japan
| | - Miki Bundo
- Department of Molecular Brain Science, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan; Laboratory for Molecular Dynamics of Mental Disorders, RIKEN Brain Science Institute, Wako, Saitama, Japan.
| | - Kazuya Iwamoto
- Department of Molecular Brain Science, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan; Laboratory for Molecular Dynamics of Mental Disorders, RIKEN Brain Science Institute, Wako, Saitama, Japan.
| |
Collapse
|
46
|
Fan Y, Yuan Y, Xiong M, Jin M, Zhang D, Yang D, Liu C, Petersen RB, Huang K, Peng A, Zheng L. Tet1 deficiency exacerbates oxidative stress in acute kidney injury by regulating superoxide dismutase. Theranostics 2023; 13:5348-5364. [PMID: 37908721 PMCID: PMC10614682 DOI: 10.7150/thno.87416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 09/14/2023] [Indexed: 11/02/2023] Open
Abstract
Rationale: Increased methylation of key genes has been observed in kidney diseases, suggesting that the ten-eleven translocation (Tet) methyl-cytosine dioxygenase family as well as 5mC oxidation may play important roles. As a member of the Tet family, the role of Tet1 in acute kidney injury (AKI) remains unclear. Methods: Tet1 knockout mice, with or without tempol treatment, a scavenger of reactive oxygen species (ROS), were challenged with ischemia and reperfusion (I/R) injury or unilateral ureteral obstruction (UUO) injury. RNA-sequencing, Western blotting, qRT-PCR, bisulfite sequencing, chromatin immunoprecipitation, immunohistochemical staining, and dot blot assays were performed. Results: Tet1 expression was rapidly upregulated following I/R or UUO injury. Moreover, Tet1 knockout mice showed increased renal injury and renal cell death, increased ROS accumulation, G2/M cell cycle arrest, inflammation, and fibrosis. Severe renal damage in injured Tet1 knockout mice was alleviated by tempol treatment. Mechanistically, Tet1 reduced the 5mC levels in an enzymatic activity-dependent manner on the promoters of Sod1 and Sod2 to promote their expression, thus lowering injury-induced excessive ROS and reducing I/R or UUO injury. Conclusions: Tet1 plays an important role in the development of AKI by promoting SOD expression through a DNA demethylase-dependent mechanism.
Collapse
Affiliation(s)
- Yu Fan
- Hubei Key Laboratory of Cell Homeostasis, Frontier Science Center for Immunology and Metabolism, College of Life Sciences, Wuhan University, Wuhan, China, 430072
| | - Yangmian Yuan
- Hubei Key Laboratory of Cell Homeostasis, Frontier Science Center for Immunology and Metabolism, College of Life Sciences, Wuhan University, Wuhan, China, 430072
| | - Mingrui Xiong
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China, 430030
| | - Muchuan Jin
- Hubei Key Laboratory of Cell Homeostasis, Frontier Science Center for Immunology and Metabolism, College of Life Sciences, Wuhan University, Wuhan, China, 430072
| | - Donge Zhang
- Department of Pharmacy, The Third Hospital of Wuhan and Tongren Hospital of Wuhan University, Wuhan, China, 430070
| | - Dong Yang
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China, 430030
| | - Chengyu Liu
- Department of Transfusion Medicine, Wuhan Hospital of Traditional Chinese and Western Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China, 430030
| | - Robert B. Petersen
- Foundational Sciences, Central Michigan University College of Medicine, Mount Pleasant, MI, USA, 48858
| | - Kun Huang
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China, 430030
| | - Anlin Peng
- Department of Pharmacy, The Third Hospital of Wuhan and Tongren Hospital of Wuhan University, Wuhan, China, 430070
| | - Ling Zheng
- Hubei Key Laboratory of Cell Homeostasis, Frontier Science Center for Immunology and Metabolism, College of Life Sciences, Wuhan University, Wuhan, China, 430072
| |
Collapse
|
47
|
Kuehner JN, Walia NR, Seong R, Li Y, Martinez-Feduchi P, Yao B. Social defeat stress induces genome-wide 5mC and 5hmC alterations in the mouse brain. G3 (BETHESDA, MD.) 2023; 13:jkad114. [PMID: 37228107 PMCID: PMC10411578 DOI: 10.1093/g3journal/jkad114] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 02/13/2023] [Accepted: 05/16/2023] [Indexed: 05/27/2023]
Abstract
Stress is adverse experience that require constant adaptation to reduce the emotional and physiological burden, or "allostatic load", of an individual. Despite their everyday occurrence, a subpopulation of individuals is more susceptible to stressors, while others remain resilient with unknown molecular signatures. In this study, we investigated the contribution of the DNA modifications, 5-methylcytosine (5mC) and 5-hydroxymethylcytosine (5hmC), underlying the individual differences in stress susceptibility and resilience. Genome-wide 5mC and 5hmC profiles from 3- and 6-month adult male mice that underwent various durations of social defeat were generated. In 3-month animals, 5mC and 5hmC work in parallel and do not distinguish between stress-susceptible and resilient phenotypes, while in 6-month animals, 5mC and 5hmC show distinct enrichment patterns. Acute stress responses may epigenetically "prime" the animals to either increase or decrease their predisposition to depression susceptibility. In support of this, re-exposure studies reveal that the enduring effects of social defeat affect differential biological processes between susceptible and resilient animals. Finally, the stress-induced 5mC and 5hmC fluctuations across the acute-chronic-longitudinal time course demonstrate that the negative outcomes of chronic stress do not discriminate between susceptible and resilient animals. However, resilience is more associated with neuroprotective processes while susceptibility is linked to neurodegenerative processes. Furthermore, 5mC appears to be responsible for acute stress response, whereas 5hmC may function as a persistent and stable modification in response to stress. Our study broadens the scope of previous research offering a comprehensive analysis of the role of DNA modifications in stress-induced depression.
Collapse
Affiliation(s)
- Janise N Kuehner
- Department of Human Genetics, Emory University School of Medicine, 615 Michael Street, Atlanta, GA 30322, USA
| | - Nevin R Walia
- Department of Human Genetics, Emory University School of Medicine, 615 Michael Street, Atlanta, GA 30322, USA
| | - Rachel Seong
- Department of Human Genetics, Emory University School of Medicine, 615 Michael Street, Atlanta, GA 30322, USA
| | - Yangping Li
- Department of Human Genetics, Emory University School of Medicine, 615 Michael Street, Atlanta, GA 30322, USA
| | - Paula Martinez-Feduchi
- Department of Human Genetics, Emory University School of Medicine, 615 Michael Street, Atlanta, GA 30322, USA
| | - Bing Yao
- Department of Human Genetics, Emory University School of Medicine, 615 Michael Street, Atlanta, GA 30322, USA
| |
Collapse
|
48
|
Sun JKL, Wong GCN, Chow KHM. Cross-talk between DNA damage response and the central carbon metabolic network underlies selective vulnerability of Purkinje neurons in ataxia-telangiectasia. J Neurochem 2023; 166:654-677. [PMID: 37319113 DOI: 10.1111/jnc.15881] [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: 04/30/2023] [Revised: 05/30/2023] [Accepted: 06/01/2023] [Indexed: 06/17/2023]
Abstract
Cerebellar ataxia is often the first and irreversible outcome in the disease of ataxia-telangiectasia (A-T), as a consequence of selective cerebellar Purkinje neuronal degeneration. A-T is an autosomal recessive disorder resulting from the loss-of-function mutations of the ataxia-telangiectasia-mutated ATM gene. Over years of research, it now becomes clear that functional ATM-a serine/threonine kinase protein product of the ATM gene-plays critical roles in regulating both cellular DNA damage response and central carbon metabolic network in multiple subcellular locations. The key question arises is how cerebellar Purkinje neurons become selectively vulnerable when all other cell types in the brain are suffering from the very same defects in ATM function. This review intended to comprehensively elaborate the unexpected linkages between these two seemingly independent cellular functions and the regulatory roles of ATM involved, their integrated impacts on both physical and functional properties, hence the introduction of selective vulnerability to Purkinje neurons in the disease will be addressed.
Collapse
Affiliation(s)
- Jacquelyne Ka-Li Sun
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong
| | - Genper Chi-Ngai Wong
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong
| | - Kim Hei-Man Chow
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong
- Gerald Choa Neuroscience Institute, The Chinese University of Hong Kong, Hong Kong
- Nexus of Rare Neurodegenerative Diseases, The Chinese University of Hong Kong, Hong Kong
| |
Collapse
|
49
|
Neofytou C, Backlund A, Blomgren K, Hermanson O. Irradiation and lithium treatment alter the global DNA methylation pattern and gene expression underlying a shift from gliogenesis towards neurogenesis in human neural progenitors. Transl Psychiatry 2023; 13:258. [PMID: 37443041 DOI: 10.1038/s41398-023-02560-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 07/01/2023] [Accepted: 07/04/2023] [Indexed: 07/15/2023] Open
Abstract
Central nervous system (CNS) tumors account for almost a third of pediatric cancers and are the largest contributor to cancer-related death in children. Cranial radiation therapy (CRT) is, often in combination with chemotherapy and surgery, effective in the treatment of high-grade childhood brain cancers, but it has been associated with late complications in 50-90% of survivors, such as decline in cognition and mood, decreased social competence, and fatigue. A leading hypothesis to explain the decline in cognition, at least partially, is injury to the neural stem and progenitor cells (NSPCs), which leads to apoptosis and altered fate choice, favoring gliogenesis over neurogenesis. Hence, treatments harnessing neurogenesis are of great relevance in this context. Lithium, a well-known mood stabilizer, has neuroprotective and antitumor effects and has been found to reverse irradiation-induced damage in rodents, at least in part by regulating the expression of the glutamate decarboxylase 2 gene (Gad2) via promoter demethylation in rat NSPCs. Additionally, lithium was shown to rescue irradiation-induced cognitive defects in mice. Here, we show that irradiation (IR) alone or in combination with lithium chloride (LiCl) caused major changes in gene expression and global DNA methylation in iPSC-derived human NSPCs (hNSPCs) compared to untreated cells, as well as LiCl-only-treated cells. The pattern of DNA methylation changes after IR-treatment alone was stochastic and observed across many different gene groups, whereas differences in DNA methylation after LiCl-treatment of irradiated cells were more directed to specific promoters of genes, including genes associated with neurogenesis, for example GAD2. Interestingly, IR and IR + LiCl treatment affected the promoter methylation and expression of several genes encoding factors involved in BMP signaling, including the BMP antagonist gremlin1. We propose that lithium in addition to promoting neuronal differentiation, also represses glial differentiation in hNSPCs with DNA methylation regulation being a key mechanism of action.
Collapse
Affiliation(s)
- Christina Neofytou
- Department of Neuroscience, Karolinska Institutet, 171 77, Stockholm, Sweden.
| | - Alexandra Backlund
- Department of Neuroscience, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Klas Blomgren
- Department of Women's and Children's Health, 171 77, Stockholm, Sweden
- Pediatric Oncology, Karolinska University Hospital, 171 64, Stockholm, Sweden
| | - Ola Hermanson
- Department of Neuroscience, Karolinska Institutet, 171 77, Stockholm, Sweden.
| |
Collapse
|
50
|
Impey S, Pelz C, Riparip LK, Tafessu A, Fareh F, Zuloaga DG, Marzulla T, Stewart B, Rosi S, Turker MS, Raber J. Postsynaptic density radiation signature following space irradiation. Front Physiol 2023; 14:1215535. [PMID: 37440997 PMCID: PMC10334289 DOI: 10.3389/fphys.2023.1215535] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 06/14/2023] [Indexed: 07/15/2023] Open
Abstract
Introduction: The response of the brain to space radiation is an important concern for astronauts during space missions. Therefore, we assessed the response of the brain to 28Si ion irradiation (600 MeV/n), a heavy ion present in the space environment, on cognitive performance and whether the response is associated with altered DNA methylation in the hippocampus, a brain area important for cognitive performance. Methods: We determined the effects of 28Si ion irradiation on object recognition, 6-month-old mice irradiated with 28Si ions (600 MeV/n, 0.3, 0.6, and 0.9 Gy) and cognitively tested two weeks later. In addition, we determined if those effects were associated with alterations in hippocampal networks and/or hippocampal DNA methylation. Results: At 0.3 Gy, but not at 0.6 Gy or 0.9 Gy, 28Si ion irradiation impaired cognition that correlated with altered gene expression and 5 hmC profiles that mapped to specific gene ontology pathways. Comparing hippocampal DNA hydroxymethylation following proton, 56Fe ion, and 28Si ion irradiation revealed a general space radiation synaptic signature with 45 genes that are associated with profound phenotypes. The most significant categories were glutamatergic synapse and postsynaptic density. Discussion: The brain's response to space irradiation involves novel excitatory synapse and postsynaptic remodeling.
Collapse
Affiliation(s)
- Soren Impey
- Department of Pediatrics, Oregon Stem Cell Center, Oregon Health and Science University, Portland, OR, United States
- Dow Neuroscience Laboratories, Department of Cell and Developmental Biology, Legacy Research Institute, Legacy Health Systems, Oregon Health and Science University, Portland, OR, United States
| | - Carl Pelz
- Department of Pediatrics, Oregon Stem Cell Center, Oregon Health and Science University, Portland, OR, United States
| | - Lara-Kirstie Riparip
- Departments of Neurological Surgery and Physical Therapy and Rehabilitation Science, Brain and Spinal Injury Center, University of California, San Francisco, San Francisco, CA, United States
| | - Amanuel Tafessu
- Department of Pediatrics, Oregon Stem Cell Center, Oregon Health and Science University, Portland, OR, United States
| | - Fatema Fareh
- Department of Pediatrics, Oregon Stem Cell Center, Oregon Health and Science University, Portland, OR, United States
| | - Damian G. Zuloaga
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR, United States
| | - Tessa Marzulla
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR, United States
| | - Blair Stewart
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR, United States
| | - Susanna Rosi
- Departments of Neurological Surgery and Physical Therapy and Rehabilitation Science, Brain and Spinal Injury Center, University of California, San Francisco, San Francisco, CA, United States
| | - Mitchell S. Turker
- Department of Molecular and Medical Genetics, Oregon Institute of Occupational Health Sciences, Oregon Health and Science University, Portland, OR, United States
| | - Jacob Raber
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR, United States
- Departments of Neurology and Radiation Medicine, Division of Neuroscience ONPRC, Oregon Health and Science University, Portland, OR, United States
| |
Collapse
|