1
|
Liu X, Feng J, Guo M, Chen C, Zhao T, Sun X, Zhang Y. Resetting the aging clock through epigenetic reprogramming: Insights from natural products. Pharmacol Ther 2025; 270:108850. [PMID: 40221101 DOI: 10.1016/j.pharmthera.2025.108850] [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: 05/01/2024] [Revised: 12/04/2024] [Accepted: 04/07/2025] [Indexed: 04/14/2025]
Abstract
Epigenetic modifications play a critical role in regulating gene expression under various physiological and pathological conditions. Epigenetic modifications reprogramming is a recognized hallmark of aging and a key component of the aging clock used to differentiate between chronological and biological age. The potential for prospective diagnosis and regulatory capabilities position epigenetic modifications as an emerging drug target to extend longevity and alleviate age-related organ dysfunctions. In the past few decades, numerous preclinical studies have demonstrated the therapeutic potential of natural products in various human diseases, including aging, with some advancing to clinical trials and clinical application. This review highlights the discovery and recent advancements in the aging clock, as well as the potential use of natural products as anti-aging therapeutics by correcting disordered epigenetic reprogramming. Specifically, the focus is on the imbalance of histone modifications, alterations in DNA methylation patterns, disrupted ATP-dependent chromatin remodeling, and changes in RNA modifications. By exploring these areas, new insights can be gained into aging prediction and anti-aging interventions.
Collapse
Affiliation(s)
- Xin Liu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin 150081, China; State Key Laboratory -Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin 150081, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin 150081, China
| | - Jing Feng
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin 150081, China; State Key Laboratory -Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin 150081, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin 150081, China
| | - Madi Guo
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin 150081, China; State Key Laboratory -Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin 150081, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin 150081, China
| | - Chen Chen
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin 150081, China; State Key Laboratory -Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin 150081, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin 150081, China
| | - Tong Zhao
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin 150081, China; State Key Laboratory -Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin 150081, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin 150081, China
| | - Xiuxiu Sun
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin 150081, China; State Key Laboratory -Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin 150081, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin 150081, China
| | - Yong Zhang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin 150081, China; State Key Laboratory -Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin 150081, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin 150081, China.
| |
Collapse
|
2
|
Zhang L, Yuan J, Yao S, Wen G, An J, Jin H, Tuo B. Role of m5C methylation in digestive system tumors (Review). Mol Med Rep 2025; 31:142. [PMID: 40183387 PMCID: PMC11979572 DOI: 10.3892/mmr.2025.13507] [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: 11/26/2024] [Accepted: 03/06/2025] [Indexed: 04/05/2025] Open
Abstract
Currently, the incidence of digestive system tumors has been increasing annually, thus becoming a prevalent cause of cancer‑related mortalities. Although significant strides have been made in targeting the molecular mechanisms that underpin the development of these tumors, their treatment and prognosis still pose substantial challenges. This is primarily due to the ambiguity of early diagnostic indicators and the fact that most digestive system tumors are detected at an advanced stage. However, epigenetic modifications are capable of altering the expression of oncogenes and regulating biological processes in cancer. In recent years, the study of methylation in relation to tumor pathogenesis has become a focus of prominent research. Among the various types of methylation, 5‑methylcytosine (m5C) methylation plays a crucial role in the development of digestive system tumors and is anticipated to serve as a novel therapeutic target. However, to date, a comprehensive and systematic review concerning the role of m5C methylation in digestive system tumors is lacking. Consequently, the present study reviewed the role of m5C methylation in digestive system tumors such as esophageal cancer, gastric cancer and hepatocellular carcinoma, with the aim of providing a valuable reference for future research endeavors.
Collapse
Affiliation(s)
- Li Zhang
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Jianbo Yuan
- Department of Laboratory Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, P.R. China
| | - Shun Yao
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Guorong Wen
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Jiaxing An
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Hai Jin
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Biguang Tuo
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| |
Collapse
|
3
|
Chen Y, Zhang S, Zeng B, Lv J, Shi B, Liu Y, Zhu M, Xu C, Cao Y, Zhang B, Dai S, Li J, Li X, Sun Q, Gu J. PUS1 facilitates cell migration in clear cell renal cell carcinoma through the promotion of mRNA pseudouridylation and the stabilization of the SMOX gene. Cell Signal 2025; 130:111700. [PMID: 39993614 DOI: 10.1016/j.cellsig.2025.111700] [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: 11/20/2024] [Revised: 02/12/2025] [Accepted: 02/21/2025] [Indexed: 02/26/2025]
Abstract
Clear cell renal cell carcinoma (ccRCC) is a highly aggressive malignancy characterized by its propensity for metastasis. Pseudouridine synthase 1 (PUS1), an enzyme involved in RNA modification, has been implicated in cancer progression, but its role in ccRCC remains unclear. This study investigates PUS1's role in ccRCC and evaluates its potential as a prognostic biomarker and therapeutic target. We assessed PUS1 expression in 541 ccRCC samples from The Cancer Genome Atlas (TCGA) and utilized Receiver Operating Characteristic (ROC) analyses to determine its prognostic value. High PUS1 levels were associated with reduced overall and disease-specific survival in ccRCC patients. Functional assays demonstrated that PUS1 promotes cell migration by stabilizing SMOX mRNA via pseudouridylation. The transcription factor USF1 regulates PUS1 expression by binding to its promoter region, enhancing its transcriptional activity. PUS1 silencing in ccRCC cell lines significantly reduced cell migration, while its overexpression had the opposite effect. These findings implicate the USF1-PUS1-SMOX signaling axis in regulating the migratory abilities of ccRCC cells. In conclusion, PUS1 plays a pivotal role in ccRCC progression and serves as an independent prognostic biomarker. Targeting PUS1 may offer promising therapeutic strategies for ccRCC patients. Further research is needed to explore its function in other cancers and to clarify its underlying molecular mechanisms.
Collapse
Affiliation(s)
- Yao Chen
- Department of Urology, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Xinhua District, Shijiazhuang, Hebei Province 050061, China
| | - Sujing Zhang
- Department of Nuclear Medicine, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Xinhua District, Shijiazhuang, Hebei Province 050061, China
| | - Bowen Zeng
- Department of Urology, The Affiliated Hospital of Sergeant School of Army Medical University, No. 10, Dajing Street, Xinhua District, Shijiazhuang, Hebei Province 050047, China
| | - Jingwei Lv
- Hebei Medical University, No. 361, Zhongshan East Road, Shijiazhuang, Hebei Province 050017, China
| | - Bei Shi
- Department of Urology, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Xinhua District, Shijiazhuang, Hebei Province 050061, China
| | - Yuepeng Liu
- Department of Urology, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Xinhua District, Shijiazhuang, Hebei Province 050061, China
| | - Meng Zhu
- Department of Urology, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Xinhua District, Shijiazhuang, Hebei Province 050061, China
| | - Chao Xu
- Department of Urology, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Xinhua District, Shijiazhuang, Hebei Province 050061, China
| | - Yilong Cao
- Department of Urology, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Xinhua District, Shijiazhuang, Hebei Province 050061, China
| | - Bowei Zhang
- Department of Urology, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Xinhua District, Shijiazhuang, Hebei Province 050061, China
| | - Shengtao Dai
- Department of Urology, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Xinhua District, Shijiazhuang, Hebei Province 050061, China
| | - Jiehan Li
- Department of Urology, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Xinhua District, Shijiazhuang, Hebei Province 050061, China
| | - Xiaoling Li
- Department of Urology, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Xinhua District, Shijiazhuang, Hebei Province 050061, China
| | - Qingyun Sun
- Department of Urology, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Xinhua District, Shijiazhuang, Hebei Province 050061, China
| | - Junfei Gu
- Department of Urology, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Xinhua District, Shijiazhuang, Hebei Province 050061, China.
| |
Collapse
|
4
|
Chaudhary JK, Danga AK, Kumari A, Bhardwaj A, Rath PC. Role of Stem Cells in Ageing and Age-related Diseases. Mech Ageing Dev 2025:112069. [PMID: 40324541 DOI: 10.1016/j.mad.2025.112069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 04/30/2025] [Accepted: 05/01/2025] [Indexed: 05/07/2025]
Abstract
Stem cell functions and ageing are deeply interconnected, continually influencing each other in multiple ways. Stem cells play a vital role in organ maintenance, regeneration, and homeostasis, all of which decline over time due to gradual reduction in their self-renewal, differentiation, and growth factor secretion potential. The functional decline is attributed to damaging extrinsic environmental factors and progressively worsening intrinsic genetic and biochemical processes. These ageing-associated deteriorative changes have been extensively documented, paving the way for the discovery of novel biomarkers of ageing for detection, diagnosis, and treatment of age-related diseases. Age-dependent changes in adult stem cells include numerical decline, loss of heterogeneity, reduced self-renewal and differentiation, leading to a drastic reduction in regenerative potential, and thereby drive the ageing process. Conversely, ageing also adversely alters the stem cell niche, disrupting the molecular pathways underlying stem cell homing, self-renewal, differentiation, and growth factor secretion, all of which are critical for tissue repair and regeneration. A holistic understanding of these molecular mechanisms, through empirical research and clinical trials, is essential for designing targeted therapies to modulate ageing and improve health parameters in older individuals.
Collapse
Affiliation(s)
- Jitendra Kumar Chaudhary
- Molecular Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi-110067, India; Department of Zoology, Shivaji College, University of Delhi, New Delhi-110027, India.
| | - Ajay Kumar Danga
- Molecular Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi-110067, India; National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi - 110067, India.
| | - Anita Kumari
- Molecular Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi-110067, India.
| | - Akshay Bhardwaj
- Global Research Alliances, Ashoka University, Rajiv Gandhi Education City, Sonepat, Haryana 131029, India.
| | - Pramod C Rath
- Molecular Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi-110067, India.
| |
Collapse
|
5
|
Tsai YL, Wolf EJ, Fluke KA, Fuchs RT, Dai N, Johnson SR, Sun Z, Elkins L, Burkhart BW, Santangelo TJ, Corrêa IR. Comprehensive nucleoside analysis of archaeal RNA modification profiles reveals an m 7G in the conserved P loop of 23S rRNA. Cell Rep 2025; 44:115471. [PMID: 40131932 DOI: 10.1016/j.celrep.2025.115471] [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: 11/21/2024] [Revised: 01/17/2025] [Accepted: 03/05/2025] [Indexed: 03/27/2025] Open
Abstract
Extremophilic Archaea employ diverse RNA modifications for survival. Our understanding of the modified nucleosides and their functions in Archaea is far from complete. Here, we establish an extensive profile of nucleoside modifications in thermophilic and mesophilic Archaea. Through liquid chromatography-tandem mass spectrometry (LC-MS/MS) and rigorous non-coding RNA depletion, we identify four previously unannotated modifications in archaeal mRNA. Nucleoside analysis conducted on total, large, small, and mRNA-enriched subfractions of hyperthermophile Thermococcus kodakarensis reveals modifications whose relative abundance is dynamically responsive to growth temperatures. To predict archaeal RNA-modifying enzymes, we leverage open-access databases to compare putative functional domains with previously annotated enzymes. Our approach leads to the discovery of a methyltransferase responsible for the installation of m7G in the P loop of 23S rRNA peptidyl transferase center in T. kodakarensis. The methyltransferase activity is confirmed in vitro with synthetic substrates and in vivo by assessing the presence of the m7G modification in a genetic deletion strain.
Collapse
MESH Headings
- RNA, Ribosomal, 23S/metabolism
- RNA, Ribosomal, 23S/chemistry
- RNA, Ribosomal, 23S/genetics
- RNA, Archaeal/metabolism
- RNA, Archaeal/genetics
- RNA, Archaeal/chemistry
- Thermococcus/genetics
- Thermococcus/metabolism
- Nucleosides/metabolism
- Methyltransferases/metabolism
- RNA Processing, Post-Transcriptional
Collapse
Affiliation(s)
| | - Eric J Wolf
- New England Biolabs Inc., Beverly, MA 01915, USA
| | - Kristin A Fluke
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA
| | - Ryan T Fuchs
- New England Biolabs Inc., Beverly, MA 01915, USA
| | - Nan Dai
- New England Biolabs Inc., Beverly, MA 01915, USA
| | | | - Zhiyi Sun
- New England Biolabs Inc., Beverly, MA 01915, USA
| | - Liam Elkins
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA
| | - Brett W Burkhart
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA
| | - Thomas J Santangelo
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA
| | | |
Collapse
|
6
|
Hou B, Jia G, Li Z, Jiang Y, Chen Y, Li X. Discovery of hydrazide-based PI3K/HDAC dual inhibitors with enhanced pro-apoptotic activity in lymphoma cells. Eur J Med Chem 2025; 292:117658. [PMID: 40300459 DOI: 10.1016/j.ejmech.2025.117658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 04/08/2025] [Accepted: 04/18/2025] [Indexed: 05/01/2025]
Abstract
PI3K and HDAC are concurrently upregulated in a variety of cancers, and simultaneous inhibition of PI3K and HDAC may synergistically inhibit tumor proliferation and induce apoptosis, providing a rationale for the study of dual-target PI3K/HDAC inhibitors. In this study, we rationally designed and synthesized a series of novel PI3K/HDAC dual-target inhibitors by combining the morpholino-triazine pharmacophore of PI3K inhibitor ZSTK474 with the hydrazide moiety of HDAC1-3 selective inhibitor 11h. Representative compound 31f possessed both PI3K (IC50 = 2.5-80.5 nM for PI3Kα, β, γ, and δ) and HDAC1-3 inhibitory activities (IC50 = 1.9-75.5 nM for HDAC1-3). 31f showed potent antiproliferative activity against a variety of tumor cell lines. Meanwhile, we designed and synthesized tool molecule 39a, a HDAC inhibitor structurally similar to 31f. In the mantle cell lymphoma Jeko-1 cell line, 31f showed significantly greater efficacy than the single inhibitors in inducing apoptosis. In conclusion, this study provided insights into the development of novel hydrazide-based dual HDAC/PI3K inhibitors.
Collapse
Affiliation(s)
- Baogeng Hou
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
| | - Geng Jia
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
| | - Zhongqiang Li
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
| | - Yuqi Jiang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
| | - Yuxin Chen
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
| | - Xiaoyang Li
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Marine Biomedical Research Institute of Qingdao, Qingdao, Shandong 266071, China.
| |
Collapse
|
7
|
Xu H, Zhao Q, Cai D, Chen X, Zhou X, Gao Y, Wu J, Yuan S, Li D, Zhang R, Peng W, Li G, Nan A. o8G-modified circKIAA1797 promotes lung cancer development by inhibiting cuproptosis. J Exp Clin Cancer Res 2025; 44:110. [PMID: 40176113 PMCID: PMC11963662 DOI: 10.1186/s13046-025-03365-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: 01/04/2025] [Accepted: 03/13/2025] [Indexed: 04/04/2025] Open
Abstract
BACKGROUND Lung cancer is a serious threat to human life and health, but effective screening and treatment methods are lacking. Circular RNAs (circRNAs) have important biological functions and are closely related to tumour development. Some studies have shown that the 8-oxo-7,8-dihydroguanosine (o8G) modification plays a key role in the disease process, but the effect of the o8G modification on circRNAs has not been elucidated. Moreover, cuproptosis is a novel mode of cell death in which copper ions directly promote protein aggregation and the disruption of cellular metabolic pathways. The present study revealed that the o8G modification of circKIAA1797 occurs and promotes lung cancer development by inhibiting cuproptosis, which provides new perspectives for epitranscriptomic studies and the development of novel therapeutic approaches for lung cancer. METHODS circRNA differential expression profiles in lung cancer were revealed via RNA high-throughput sequencing, and circKIAA1797 expression in lung cancer cell lines and tissues was detected using qPCR. Experiments such as o8G RNA immunoprecipitation (o8G RIP) and crosslinking immunoprecipitation (CLIP) were performed to explore the presence of o8G on circKIAA1797. The regulation of circKIAA1797 by the o8G reader Y-box binding protein 1 (YBX1) was explored using nuclear-cytoplasmic fractionation, actinomycin D (Act D) stability experiments and other experiments. circKIAA1797 silencing and overexpression systems were constructed for in vivo and in vitro experiments to study the role of circKIAA1797 in lung cancer development. Tagged RNA affinity purification (TRAP), RNA immunoprecipitation (RIP), coimmunoprecipitation (Co-IP), and immunofluorescence (IF) staining were subsequently conducted to reveal the molecular mechanism by which circKIAA1797 regulates cuproptosis and promotes lung cancer development. RESULTS This study is the first to reveal the presence of o8G on circKIAA1797 and that YBX1 is a reader that recognises ROS-induced circKIAA1797 o8G modifications and increases the stability and cytoplasmic expression of circKIAA1797. circKIAA1797, which is associated with the tumour stage and prognosis, has been shown to significantly promote the biological function of lung cancer development both in vivo and in vitro. This study revealed that circKIAA1797 inhibits intracellular cuproptosis by binding to the ferredoxin 1 (FDX1) mRNA, decreasing FDX1 mRNA stability, inhibiting FDX1 expression, and binding to the signal transducer and activator of transcription 1 (STAT1) protein and inhibiting lipoyltransferase 1 (LIPT1) transcription; moreover, circKIAA1797 promotes the closure of the mitochondrial permeability transition pore (mPTP), inhibits cuproptosis, and ultimately promotes lung cancer development. CONCLUSIONS This study revealed the presence of the o8G modification in circKIAA1797, which plays an important role in the development of lung cancer. circKIAA1797 can inhibit cuproptosis by inhibiting key cuproptosis proteins and promoting mPTP closure, ultimately promoting the development of lung cancer. This study provides not only a new theoretical basis for an in-depth understanding of the molecular mechanisms of lung cancer development but also a potential target for lung cancer treatment.
Collapse
Affiliation(s)
- Haotian Xu
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Qingyun Zhao
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Dunyu Cai
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Xingcai Chen
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Xiaodong Zhou
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Yihong Gao
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Jiaxi Wu
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Shengyi Yuan
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Deqing Li
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Ruirui Zhang
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Wenyi Peng
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Gang Li
- School of Public Health, Guangxi Medical University, Nanning, 530021, China.
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China.
| | - Aruo Nan
- School of Public Health, Guangxi Medical University, Nanning, 530021, China.
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China.
| |
Collapse
|
8
|
Hoehn SJ, Krul SE, Seth SK, Crespo-Hernández CE. Structure-Photophysical Property Relationships in Noncanonical and Synthetic Nucleobases. Annu Rev Phys Chem 2025; 76:539-564. [PMID: 39952631 DOI: 10.1146/annurev-physchem-082423-022427] [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] [Indexed: 02/17/2025]
Abstract
This review provides focused coverage of the photophysical properties of noncanonical and synthetic nucleobases reported over the past decade. It emphasizes key research findings and physical insights gathered for prebiotic and fluorescent nucleobase analogs, sulfur- and selenium-substituted nucleobases, aza-substituted nucleobases, epigenetic nucleobases and their oxidation products, and nucleobases utilized for expanding DNA/RNA to reveal central structure-photophysical property relationships. Further research and development in this emerging field, coupled with machine learning methods, will enable the effective harnessing of nucleobases' modifications for applications in biotechnology, biomedicine, therapeutics, and even the creation of live semisynthetic organisms.
Collapse
Affiliation(s)
- Sean J Hoehn
- Department of Chemistry, Case Western Reserve University, Cleveland, Ohio, USA;
| | - Sarah E Krul
- Department of Chemistry, Case Western Reserve University, Cleveland, Ohio, USA;
| | - Sourav Kanti Seth
- Department of Chemistry, Case Western Reserve University, Cleveland, Ohio, USA;
| | | |
Collapse
|
9
|
Fakeri M, Shakoul F, Yaghoubi SM, Koulaeizadeh S, Haghi M. Comprehensive insights into circular RNAs, miRNAs, and lncRNAs as biomarkers in retinoblastoma. Ophthalmic Genet 2025; 46:122-132. [PMID: 39849678 DOI: 10.1080/13816810.2025.2456607] [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: 05/03/2024] [Revised: 12/21/2024] [Accepted: 01/16/2025] [Indexed: 01/25/2025]
Abstract
Retinoblastoma (RB) is a common and potentially lethal cancer that primarily affects young children worldwide, with survival rates significantly varying between high- and low-income countries. This review aims to identify essential diagnostic markers for early diagnosis by investigating the molecular pathways associated with RB. The prevalence of RB cases is notably concentrated in Asia and Africa, contributing to a global survival rate estimate of less than 30%. Current management strategies involve complex, individualized treatment plans that consider cultural nuances, genetic abnormalities, staging, and the availability of medical resources. Recent studies suggest that circular RNAs (circRNAs) may serve as predictive and diagnostic biomarkers in the etiology of RB. This review examines the roles of long non-coding RNAs (lncRNAs), microRNAs (miRNAs), and circRNAs in RB, with the goal of improving survival rates, particularly in low- and middle-income countries. A deeper understanding of the molecular pathways of RB may facilitate the development of personalized treatment plans and targeted therapies. Elevated expression of circRNAs has been observed in most patient cases, and studies indicate that reducing specific circRNA production can inhibit tumor cell development and progression. Investigating the roles and mechanisms of circular RNAs in RB holds promise for future treatment approaches.
Collapse
Affiliation(s)
- Mahsa Fakeri
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Fatemeh Shakoul
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | | | - Shabnam Koulaeizadeh
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Mehdi Haghi
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| |
Collapse
|
10
|
Wang H, Huang J, Fang X, Liu M, Fan X, Li Y. Advances in next-generation sequencing (NGS) applications in drug discovery and development. Expert Opin Drug Discov 2025; 20:537-550. [PMID: 40099494 DOI: 10.1080/17460441.2025.2481262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 02/27/2025] [Accepted: 03/14/2025] [Indexed: 03/20/2025]
Abstract
INTRODUCTION Drug discovery is a complex and multifaceted process driven by scientific innovation and advanced technologies. Next-Generation Sequencing (NGS) platforms, encompassing both short-read and long-read technologies, have revolutionized the field by enabling the high-throughput and cost-effective analysis of DNA and RNA molecules. Continuous advancements in NGS-based technologies have enabled their seamless integration across preclinical and clinical workflows in drug discovery, encompassing early-stage drug target identification, candidate selection, genetically stratified clinical trials, and pharmacogenetic studies. AREA COVERED This review provides an overview of the current and potential applications of NGS-based technologies in drug discovery and development process, including their roles in novel drug target identification, high-throughput screening, clinical trials, and clinical medication studies. The review is based on literature retrieval from the PubMed and Web of Science databases between 2018 and 2024. EXPERT OPINION As technologies advance rapidly, NGS enhances accuracy and generates vast datasets. These datasets are extensively integrated with other heterogeneous data in systems biology and are mined using machine learning to extract significant insights, thereby driving progress in drug discovery.
Collapse
Affiliation(s)
- Huihong Wang
- Pharmaceutical Department, Chongqing University Three Gorges Hospital, Chongqing University, Chongqing, P. R. China
| | - Jiale Huang
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Innovative Drug Research Center, School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| | - Xianfu Fang
- Pharmaceutical Department, Chongqing University Three Gorges Hospital, Chongqing University, Chongqing, P. R. China
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Innovative Drug Research Center, School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| | - Mengyao Liu
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Innovative Drug Research Center, School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| | - Xiaohong Fan
- Pharmaceutical Department, Chongqing University Three Gorges Hospital, Chongqing University, Chongqing, P. R. China
| | - Yizhou Li
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Innovative Drug Research Center, School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| |
Collapse
|
11
|
Xiong Q, Zhang Y, Zheng Y, Zhu Q. Regulation and application of m 6A modification in tumor immunity. SCIENCE CHINA. LIFE SCIENCES 2025; 68:974-993. [PMID: 39648245 DOI: 10.1007/s11427-024-2648-0] [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: 03/02/2024] [Accepted: 06/11/2024] [Indexed: 12/10/2024]
Abstract
The m6A modification is an RNA modification that impacts various processes of RNA molecules, including transcription, splicing, stability, and translation. Recently, researchers have discovered that the presence of m6A modification can influence the interaction between tumor cells and immune cells and also play a role in regulating the expression of immune response-related genes. Additionally, m6A modification is intricately involved in the regulation of tumor immune evasion and drug resistance. Specifically, certain tumor cells can manipulate the gene expression through m6A modification to evade immune system attacks. Therefore, it might be possible to enhance tumor immune surveillance and improve the effectiveness of immune-based therapies by manipulating m6A modification. This review systematically discusses the role of m6A modification in tumor immunity, specifically highlighting its regulation of immune cells and immune-related genes in tumor cells. Furthermore, we explore the potential of m6A modification inhibitors as anti-cancer therapies and the significance of m6A regulatory factors in predicting the efficacy of tumor immune therapy.
Collapse
Affiliation(s)
- Qunli Xiong
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yaguang Zhang
- Laboratory of Gastrointestinal Tumor Epigenetics and Genomics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ying Zheng
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qing Zhu
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
12
|
Coopman E, D'Hert S, Rademakers R, De Coster W. Methylmap: visualization of modified nucleotides for large cohort sizes. BMC Bioinformatics 2025; 26:91. [PMID: 40140766 PMCID: PMC11948879 DOI: 10.1186/s12859-025-06106-3] [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: 01/08/2025] [Accepted: 03/07/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND Over the years, there has been growing interest in epigenetics, where nucleotide modifications are increasingly recognized for their roles in health and disease. Understanding methylation patterns at the nucleotide level has become pivotal for advancing this field. However, visualizing these modifications, particularly in cohorts of more than a few individuals, remains a challenge. RESULTS Here, we present methylmap, a tool developed to visualize modified nucleotide frequencies for regions of interest, specifically optimized for cohort sizes with more than a few individuals. Furthermore, methylmap features the visualization of the haplotype-specific methylation status of 226 individuals of the 1000 Genomes Project ONT Sequencing Consortium, sequenced using the Oxford Nanopore Technologies PromethION. This resource provides the research community with a comprehensive and complete overview of genome-wide methylation patterns. CONCLUSIONS Methylmap offers an easy-to-use platform to facilitate epigenetic research. It is available both as a web application at https://methylmap.bioinf.be and as a command-line tool through Bioconda and PyPI. As such, we provide a valuable resource for advancing the understanding of epigenetic modifications in health and disease.
Collapse
Affiliation(s)
- Elise Coopman
- VIB Center for Molecular Neurology, VIB, Universiteitsplein 1, 2610, Wilrijk, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Svenn D'Hert
- VIB Center for Molecular Neurology, VIB, Universiteitsplein 1, 2610, Wilrijk, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Rosa Rademakers
- VIB Center for Molecular Neurology, VIB, Universiteitsplein 1, 2610, Wilrijk, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Wouter De Coster
- VIB Center for Molecular Neurology, VIB, Universiteitsplein 1, 2610, Wilrijk, Antwerp, Belgium.
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
13
|
PANG GUANTING, LI YAOHAN, SHI QIWEN, TIAN JINGKUI, LOU HANMEI, FENG YUE. Omics sciences for cervical cancer precision medicine from the perspective of the tumor immune microenvironment. Oncol Res 2025; 33:821-836. [PMID: 40191729 PMCID: PMC11964870 DOI: 10.32604/or.2024.053772] [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: 05/10/2024] [Accepted: 08/01/2024] [Indexed: 04/09/2025] Open
Abstract
Immunotherapies have demonstrated notable clinical benefits in the treatment of cervical cancer (CC). However, the development of therapeutic resistance and diverse adverse effects in immunotherapy stem from complex interactions among biological processes and factors within the tumor immune microenvironment (TIME). Advanced omic technologies offer novel insights into a more expansive and thorough layer of the TIME. Furthermore, integrating multidimensional omics within the frameworks of systems biology and computational methodologies facilitates the generation of interpretable data outputs to characterize the clinical and biological trajectories of tumor behavior. In this review, we present advanced omics technologies that utilize various clinical samples to address scientific inquiries related to immunotherapies for CC, highlighting their utility in identifying metastasis dissemination, recurrence risk, and therapeutic resistance in patients treated with immunotherapeutic approaches. This review elaborates on the strategy for integrating multi-omics data through artificial intelligence algorithms. Additionally, an analysis of the obstacles encountered in the multi-omics analysis process and potential avenues for future research in this domain are presented.
Collapse
Affiliation(s)
- GUANTING PANG
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China
| | - YAOHAN LI
- College of Artificial Intelligence and Big Data for Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250000, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China
| | - QIWEN SHI
- Collaborative Innovation Center for Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, 310014, China
| | - JINGKUI TIAN
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China
| | - HANMEI LOU
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China
- Department of Gynecological Oncology, Zhejiang Cancer Hospital, Hangzhou, 310022, China
| | - YUE FENG
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China
- Department of Gynecological Oncology, Zhejiang Cancer Hospital, Hangzhou, 310022, China
| |
Collapse
|
14
|
Lancaster CL, Moberg KH, Corbett AH. Post-Transcriptional Regulation of Gene Expression and the Intricate Life of Eukaryotic mRNAs. WILEY INTERDISCIPLINARY REVIEWS. RNA 2025; 16:e70007. [PMID: 40059537 PMCID: PMC11949413 DOI: 10.1002/wrna.70007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 02/17/2025] [Accepted: 02/18/2025] [Indexed: 03/29/2025]
Abstract
In recent years, there has been a growing appreciation for how regulatory events that occur either co- or post-transcriptionally contribute to the control of gene expression. Messenger RNAs (mRNAs) are extensively regulated throughout their metabolism in a precise spatiotemporal manner that requires sophisticated molecular mechanisms for cell-type-specific gene expression, which dictates cell function. Moreover, dysfunction at any of these steps can result in a variety of human diseases, including cancers, muscular atrophies, and neurological diseases. This review summarizes the steps of the central dogma of molecular biology, focusing on the post-transcriptional regulation of gene expression.
Collapse
Affiliation(s)
- Carly L. Lancaster
- Department of Biology, Emory College of Arts and Sciences, Atlanta, Georgia, USA
- Department of Cell Biology Emory University School of Medicine, Atlanta, Georgia, USA
- Graduate Program in Biochemistry, Cell and Developmental Biology, Emory University Atlanta, Georgia, USA
| | - Kenneth H. Moberg
- Department of Cell Biology Emory University School of Medicine, Atlanta, Georgia, USA
| | - Anita H. Corbett
- Department of Biology, Emory College of Arts and Sciences, Atlanta, Georgia, USA
| |
Collapse
|
15
|
Zhang D, Gan Y, Le L, Pu L. Epigenetic variation in maize agronomical traits for breeding and trait improvement. J Genet Genomics 2025; 52:307-318. [PMID: 38310944 DOI: 10.1016/j.jgg.2024.01.006] [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: 12/04/2023] [Revised: 01/28/2024] [Accepted: 01/29/2024] [Indexed: 02/06/2024]
Abstract
Epigenetics-mediated breeding (epibreeding) involves engineering crop traits and stress responses through the targeted manipulation of key epigenetic features to enhance agricultural productivity. While conventional breeding methods raise concerns about reduced genetic diversity, epibreeding propels crop improvement through epigenetic variations that regulate gene expression, ultimately impacting crop yield. Epigenetic regulation in crops encompasses various modes, including histone modification, DNA modification, RNA modification, non-coding RNA, and chromatin remodeling. This review summarizes the epigenetic mechanisms underlying major agronomic traits in maize and identifies candidate epigenetic landmarks in the maize breeding process. We propose a valuable strategy for improving maize yield through epibreeding, combining CRISPR/Cas-based epigenome editing technology and Synthetic Epigenetics (SynEpi). Finally, we discuss the challenges and opportunities associated with maize trait improvement through epibreeding.
Collapse
Affiliation(s)
- Daolei Zhang
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China; School of Life Science, Inner Mongolia University, Hohhot, Inner Mongolia 010021, China
| | - Yujun Gan
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Liang Le
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China.
| | - Li Pu
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China.
| |
Collapse
|
16
|
Zhang Y, Song JY, Sun ZG. Exploring the impact of environmental factors on male reproductive health through epigenetics. Reprod Toxicol 2025; 132:108832. [PMID: 39778664 DOI: 10.1016/j.reprotox.2025.108832] [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: 11/04/2024] [Revised: 01/01/2025] [Accepted: 01/04/2025] [Indexed: 01/11/2025]
Abstract
Male infertility has become an increasingly severe global health issue, with its incidence significantly rising over the past few decades. This paper delves into the crucial role of epigenetics in male reproductive health, focusing particularly on the effects of DNA methylation, histone modifications, chromatin remodeling and non-coding RNAs regulation on spermatogenesis. Exposure to various environmental factors can cause sperm DNA damage, leading to epigenetic abnormalities. Among these factors, we have discussed heavy metals (including Zinc, Cadmium, Arsenic, Copper), phthalates, electromagnetic radiation, and temperature in detail. Notably, aberrations in DNA methylation are closely associated with various symptoms of male infertility, and histone modifications and chromatin remodeling are essential for sperm maturation and function. By synthesizing existing literature and experimental data, this narrative review investigates how environmental factors influence male reproductive health through epigenetic mechanisms, thus providing new theoretical foundations and practical guidelines for the early diagnosis and treatment of male infertility.
Collapse
Affiliation(s)
- Yi Zhang
- The First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, China.
| | - Jing-Yan Song
- Reproductive and Genetic Center, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China.
| | - Zhen-Gao Sun
- Reproductive and Genetic Center, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China.
| |
Collapse
|
17
|
Shen J, Ding Y. Multifaceted roles of insulin‑like growth factor 2 mRNA binding protein 2 in human cancer (Review). Mol Med Rep 2025; 31:75. [PMID: 39886962 PMCID: PMC11795254 DOI: 10.3892/mmr.2025.13441] [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: 09/19/2024] [Accepted: 01/07/2024] [Indexed: 02/01/2025] Open
Abstract
Insulin‑like growth factor 2 mRNA binding protein 2 (IGF2BP2) is an RNA binding protein that functions as an N6‑methyladenosine reader. It regulates various biological processes in human cancers by affecting the stability and expression of target RNA transcripts, including coding RNAs and non‑coding RNAs (ncRNAs). Numerous studies have shown that IGF2BP2 expression is aberrantly increased in various types of cancer and plays multifaceted roles in the development and progression of human cancers. In the present review, the clinical importance of IGF2BP2 is summarized and its involvement in the regulation of biological processes, including proliferation, metastasis, chemoresistance, metabolism, tumor immunity, stemness and cell death, in human cancers is discussed. The chemical compounds that have been developed as IGF2BP2 inhibitors are also detailed. As ncRNAs are now important potential therapeutic agents for cancer treatment, the microRNAs that have been reported to directly target and inhibit IGF2BP2 expression in cancers are also described. In summary, by reviewing the latest literature, the present study aimed to highlight the clinical importance and physiological functions of IGF2BP2 in human cancer, with a focus on the great potential of IGF2BP2 as a target for inhibitor development. The present review may inspire new ideas for future studies on IGF2BP2, which may serve as a specific therapeutic target in cancer.
Collapse
Affiliation(s)
- Jianan Shen
- Department of Pathology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu 210008, P.R. China
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 210009, P.R. China
| | - Youxiang Ding
- Department of Pathology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu 210008, P.R. China
| |
Collapse
|
18
|
Chen X, Yuan Y, Zhou F, Li L, Pu J, Jiang X. m6A RNA methylation: a pivotal regulator of tumor immunity and a promising target for cancer immunotherapy. J Transl Med 2025; 23:245. [PMID: 40022120 PMCID: PMC11871626 DOI: 10.1186/s12967-025-06221-y] [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: 09/27/2024] [Accepted: 02/11/2025] [Indexed: 03/03/2025] Open
Abstract
M6A modification is one of the most common regulatory mechanisms of gene expression in eukaryotic cells, influencing processes such as RNA splicing, degradation, stability, and protein translation. Studies have shown that m6A methylation is closely associated with tumorigenesis and progression, and it plays a key regulatory role in tumor immune responses. m6A modification participates in regulating the differentiation and maturation of immune cells, as well as related anti-tumor immune responses. In the tumor microenvironment, m6A modification can also affect immune cell recruitment, activation, and polarization, thereby promoting or inhibiting tumor cell proliferation and metastasis, and reshaping the tumor immune microenvironment. In recent years, immunotherapies for tumors, such as immune checkpoint inhibitors and adoptive cell immunotherapy, have been increasingly applied in clinical settings, achieving favorable outcomes. Targeting m6A modifications to modulate the immune system, such as using small-molecule inhibitors to target dysregulated m6A regulatory factors or inducing immune cell reprogramming, can enhance anti-tumor immune responses and strengthen immune cell recognition and cytotoxicity against tumor cells. m6A modification represents a new direction in tumor immunotherapy with promising clinical potential. This review discusses the regulatory role of m6A methylation on immune cells and tumor immune responses and explores new strategies for immunotherapy.
Collapse
Affiliation(s)
- Xi Chen
- Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650223, China
- NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, Yunnan, 650500, China
| | - Yixiao Yuan
- Department of Medicine, UF Health Cancer Center, University of Florida, Gainesville, FL, 32610, USA
| | - Fan Zhou
- Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650223, China
- NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, Yunnan, 650500, China
| | - Lihua Li
- NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, Yunnan, 650500, China
| | - Jun Pu
- Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650223, China.
- NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, Yunnan, 650500, China.
| | - Xiulin Jiang
- Department of Medicine, UF Health Cancer Center, University of Florida, Gainesville, FL, 32610, USA.
| |
Collapse
|
19
|
Al-Malsi K, Xie S, Cai Y, Mohammed N, Xie K, Lan T, Wu H. The role of lactylation in tumor growth and cancer progression. Front Oncol 2025; 15:1516785. [PMID: 39968078 PMCID: PMC11832377 DOI: 10.3389/fonc.2025.1516785] [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: 10/24/2024] [Accepted: 01/16/2025] [Indexed: 02/20/2025] Open
Abstract
Background Lactate's perception of lactate has changed over the last 30 years from a straightforward metabolic byproduct to a complex chemical with important biological activities, such as signal transduction, gluconeogenesis, and mitochondrial respiration. In addition to its metabolic contributions, lactate has far-reaching repercussions. This review highlights the role of lactate in the course of cancer by highlighting lactylation as a unique epigenetic alteration. The purpose of this review is to clarify the functions of lactate in the biology of tumors, with a particular focus on the translational potential of lactylation pathways in cancer diagnosis and treatment approaches. Methods This review summarizes research on the relationship between lactate and cancer, with an emphasis on histone lactylation, its effect on gene expression, and its influence on the tumor microenvironment. By establishing a connection between metabolic byproducts and epigenetic gene regulation, we investigated how lactylation affects immune regulation, inflammation, and cellular repair. Findings Histone lactylation, or the addition of lactate to lysine residues on histone proteins, increases transcriptional activity and facilitates the expression of genes involved in homeostasis and repair. These findings have important implications for cancer treatment. Lactylation, for example, activates genes such as Arg1, which is a hallmark of the M2 macrophage phenotype implicated in immunosuppression and tumor growth. The ability of lactate to dynamically alter gene expression is further supported by its function as a histone deacetylase(HDAC)inhibitor and its impact on histone acetylation. Its wide-ranging involvement in cellular metabolism and epigenetic control has been demonstrated by the discovery of particular lactylation sites on histones in various cell types, including cancer cells.
Collapse
Affiliation(s)
- Khulood Al-Malsi
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
- Liver Transplant Center, Transplant Center, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Hepatic AI Translation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Sinan Xie
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
- Liver Transplant Center, Transplant Center, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Hepatic AI Translation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yunshi Cai
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
- Liver Transplant Center, Transplant Center, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Hepatic AI Translation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Nader Mohammed
- Emergency Department Research, Hamad Medical Corporation, Doha, Qatar
| | - Kunlin Xie
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
- Liver Transplant Center, Transplant Center, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Hepatic AI Translation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Tian Lan
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
- Liver Transplant Center, Transplant Center, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Hepatic AI Translation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Hong Wu
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
- Liver Transplant Center, Transplant Center, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Hepatic AI Translation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
20
|
Fang H, He J, Du D, Wang X, Xu X, Lu L, Zhou Y, Wen Y, He F, Li Y, Wen H, Zhou M. Deciphering the secret codes in N 7-methylguanosine modification: Context-dependent function of methyltransferase-like 1 in human diseases. Clin Transl Med 2025; 15:e70240. [PMID: 39979979 PMCID: PMC11842222 DOI: 10.1002/ctm2.70240] [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: 09/25/2024] [Revised: 02/05/2025] [Accepted: 02/12/2025] [Indexed: 02/22/2025] Open
Abstract
N7-methylguanosine (m7G) is one of the most prevalent post-transcriptional modifications of RNA and plays a critical role in RNA translation and stability. As a pivotal m7G regulator, methyltransferase-like 1 (METTL1) is responsible for methyl group transfer during the progression of m7G modification and contributes to the structure and functional regulation of RNA. Accumulating evidence in recent years has revealed that METTL1 plays key roles in various diseases depending on its m7G RNA methyltransferase activity. Elevated levels of METTL1 are typically associated with disease development and adverse consequences. In contrast, METTL1 may act as a disease suppressor in several disorders. While the roles of m7G modifications in disease have been extensively reviewed, the critical functions of METTL1 in various types of disease and the potential targeting of METTL1 for disease treatment have not yet been highlighted. This review describes the various biological functions of METTL1, summarises recent advances in understanding its pathogenic and disease-suppressive functions and discusses the underlying molecular mechanisms. Given that METTL1 can promote or inhibit disease processes, the possibility of applying METTL1 inhibitors and agonists is further discussed, with the goal of providing novel insights for future disease diagnosis and potential intervention targets. KEY POINTS: METTL1-mediated m7G modification is crucial for various biological processes, including RNA stability, maturation and translation. METTL1 has emerged as a critical epigenetic modulator in human illnesses, with its dysregulated expression correlating with multiple diseases progression and presenting opportunities for both diagnostic biomarker development and molecular-targeted therapy. Enormous knowledge gaps persist regarding context-dependent regulatory networks of METTL1 and dynamic m7G modification patterns, necessitating mechanistic interrogation to bridge basic research with clinical translation in precision medicine.
Collapse
Affiliation(s)
- Huan Fang
- Department of GastroenterologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Jing He
- Department of Breast SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Dan Du
- Department of Medical LaboratoryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Xue Wang
- Department of Medical LaboratoryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Xinyu Xu
- Department of Medical LaboratoryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Linping Lu
- Department of GastroenterologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Yefan Zhou
- Department of Medical LaboratoryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Yangyang Wen
- Department of GastroenterologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Fucheng He
- Department of Medical LaboratoryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Yingxia Li
- Department of GastroenterologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Hongtao Wen
- Department of GastroenterologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Mingxia Zhou
- Department of GastroenterologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| |
Collapse
|
21
|
Si X, Chen X, Guo B, Liao Z, Yan X, Qi P. The role of methyltransferase-like 3 (METTL3) in immune response modulation in bivalve (Mytilus coruscus) during bacterial infection. FISH & SHELLFISH IMMUNOLOGY 2025; 157:110094. [PMID: 39716580 DOI: 10.1016/j.fsi.2024.110094] [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: 08/03/2024] [Revised: 12/17/2024] [Accepted: 12/21/2024] [Indexed: 12/25/2024]
Abstract
N6-methyladenosine (m6A) modification is a prevalent mRNA modification that regulates diverse biological processes in eukaryotes, including immune responses. While the role of m6A in mammalian immunity has been explored, its involvement in the immune defense of invertebrates, particularly marine bivalves which face constant pathogen challenges, remains largely unknown. Here, we investigated the function of methyltransferase-like 3 (METTL3), a key m6A "writer" enzyme, in the immune response of the marine bivalve Mytilus coruscus against Vibrio alginolyticus infection. M. coruscus METTL3 (McMETTL3) expression in the digestive gland increased (3-fold) after V. alginolyticus infection, coinciding with elevated m6A levels. Silencing McMETTL3 reduced both m6A levels and V. alginolyticus-induced apoptosis in digestive gland cells. In silico analysis identified a C1q-like protein family member (McC1QL) as a potential downstream target of McMETTL3, exhibiting an increase (7.2-fold) in m6A modification and an increase (1.5-fold) in expression during infection. Functional experiments confirmed that McC1QL knockdown inhibited McMETTL3-driven apoptosis (10.83 %). These findings demonstrate that METTL3 regulates apoptosis and immune responses in invertebrates via m6A modification of target genes like McC1QL. This study provides novel insights into the m6A-mediated immune regulation mechanisms in marine bivalves and may offer potential avenues for developing innovative disease control strategies in aquaculture.
Collapse
Affiliation(s)
- Xirui Si
- National Engineering Research Center of Marine Facilities Aquaculture, Marine Science and Technology College, Zhejiang Ocean University, Zhoushan, Zhejiang, 316004, China
| | - Xinglu Chen
- National Engineering Research Center of Marine Facilities Aquaculture, Marine Science and Technology College, Zhejiang Ocean University, Zhoushan, Zhejiang, 316004, China
| | - Baoying Guo
- National Engineering Research Center of Marine Facilities Aquaculture, Marine Science and Technology College, Zhejiang Ocean University, Zhoushan, Zhejiang, 316004, China
| | - Zhi Liao
- National Engineering Research Center of Marine Facilities Aquaculture, Marine Science and Technology College, Zhejiang Ocean University, Zhoushan, Zhejiang, 316004, China
| | - Xiaojun Yan
- National Engineering Research Center of Marine Facilities Aquaculture, Marine Science and Technology College, Zhejiang Ocean University, Zhoushan, Zhejiang, 316004, China
| | - Pengzhi Qi
- National Engineering Research Center of Marine Facilities Aquaculture, Marine Science and Technology College, Zhejiang Ocean University, Zhoushan, Zhejiang, 316004, China.
| |
Collapse
|
22
|
Wang G, Liang X, Wu Z, Fan B, Wang J, Zheng Q, Li D, An T. Improved biosynthesis of tyrosol by epigenetic modification-based regulation and metabolic engineering in Saccharomyces cerevisiae. J Biotechnol 2025; 398:175-182. [PMID: 39746378 DOI: 10.1016/j.jbiotec.2024.12.013] [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: 09/03/2024] [Revised: 12/06/2024] [Accepted: 12/30/2024] [Indexed: 01/04/2025]
Abstract
Aromatic amino acids and their derivatives are high value chemicals widely used in food, pharmaceutical and feed industries. Current preparation methods for aromatic amino acid products are fraught with limitations. In this study, the efficient biosynthesis of aromatic amino acid compound tyrosol was investigated by epigenetic modification-based regulation and optimization of the biosynthetic pathway of aromatic amino acids. The production of tyrosol was significantly improved by the overexpression of m6A modification writer Ime4 and reader Pho92, and the positive regulator Gcr2. Introduction of Bbxfpk and deletion of Gpp1 further improved tyrosol production. Then the feedback inhibition of the shikimate pathway was relieved by the mutants Aro4K229L and Aro7G141S. The final tyrosol producing engineered strain was constructed by the deletion of PHA2, replacement of the native promoter of ARO10 with the strong promoter PGK1p, and introduction of tyrosine decarboxylase PcAAS. In the background of m6A modification regulation, this strain ultimately produced 954.69 ± 43.72 mg/L of tyrosol, promoted by 61.7-fold in shake-flask fermentation.
Collapse
Affiliation(s)
- Guoli Wang
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Binzhou Medical University, Yantai 264003, PR China
| | - Xiqin Liang
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Binzhou Medical University, Yantai 264003, PR China
| | - Zhenke Wu
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Binzhou Medical University, Yantai 264003, PR China
| | - Bengui Fan
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Binzhou Medical University, Yantai 264003, PR China
| | - Jun Wang
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Binzhou Medical University, Yantai 264003, PR China
| | - Qiusheng Zheng
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Binzhou Medical University, Yantai 264003, PR China
| | - Defang Li
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Binzhou Medical University, Yantai 264003, PR China.
| | - Tianyue An
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Binzhou Medical University, Yantai 264003, PR China.
| |
Collapse
|
23
|
Wu Q, Fu X, Liu G, He X, Li Y, Ou C. N7-methylguanosine modification in cancers: from mechanisms to therapeutic potential. J Hematol Oncol 2025; 18:12. [PMID: 39881381 PMCID: PMC11780989 DOI: 10.1186/s13045-025-01665-7] [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: 11/27/2024] [Accepted: 01/13/2025] [Indexed: 01/31/2025] Open
Abstract
N7-methylguanosine (m7G) is an important RNA modification involved in epigenetic regulation that is commonly observed in both prokaryotic and eukaryotic organisms. Their influence on the synthesis and processing of messenger RNA, ribosomal RNA, and transfer RNA allows m7G modifications to affect diverse cellular, physiological, and pathological processes. m7G modifications are pivotal in human diseases, particularly cancer progression. On one hand, m7G modification-associated modulate tumour progression and affect malignant biological characteristics, including sustained proliferation signalling, resistance to cell death, activation of invasion and metastasis, reprogramming of energy metabolism, genome instability, and immune evasion. This suggests that they may be novel therapeutic targets for cancer treatment. On the other hand, the aberrant expression of m7G modification-associated molecules is linked to clinicopathological characteristics, including tumour staging, lymph node metastasis, and unfavourable prognoses in patients with cancer, indicating their potential as tumour biomarkers. This review consolidates the discovery, identification, detection methodologies, and functional roles of m7G modification, analysing the mechanisms by which m7G modification-associated molecules contribute to tumour development, and exploring their potential clinical applications in cancer diagnostics and therapy, thereby providing innovative strategies for tumour identification and targeted treatment.
Collapse
Affiliation(s)
- Qihui Wu
- Department of Gynecology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Xiaodan Fu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Guoqian Liu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Xiaoyun He
- Departments of Ultrasound Imaging, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| | - Yimin Li
- Department of Pathology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China.
| | - Chunlin Ou
- Department of Gynecology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
24
|
Liang Z, Liu W, Cao M, Cui J, Lan J, Ding Y, Zhang T, Yang Z. Epigenetic regulation-mediated disorders in dopamine transporter endocytosis: A novel mechanism for the pathogenesis of Parkinson's disease. Theranostics 2025; 15:2250-2278. [PMID: 39990232 PMCID: PMC11840736 DOI: 10.7150/thno.107436] [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: 11/22/2024] [Accepted: 12/30/2024] [Indexed: 02/25/2025] Open
Abstract
Mechanisms such as DNA methylation, histone modifications, and non-coding RNA regulation may impact the endocytosis of dopamine transporter (DAT) by influencing processes like neuronal survival, thereby contributing to the initiation and progression of Parkinson's Disease (PD). Some small molecule inhibitors or natural bioactive compounds have the potential to modulate epigenetic processes, thereby reversing induced pluripotent stem cells (iPSCs) reprogramming and abnormal differentiation, offering potential therapeutic effects for PD. Although no specific DNA modification enzyme directly regulates DAT endocytosis, enzymes such as DNA methyltransferases (DNMTs) may indirectly influence DAT endocytosis by regulating the expression of genes associated with this process. DNA modifications impact DAT endocytosis by modulating key signaling pathways, including the (protein kinase C) PKC and D2 receptor (D2R) pathways. Key enzymes involved in RNA modifications that influence DAT endocytosis include m6A methyltransferases and other related enzymes. This regulation impacts the synthesis and function of proteins involved in DAT endocytosis, thereby indirectly affecting the process itself. RNA modifications regulate DAT endocytosis through various indirect pathways, as well as histone modifications. Key enzymes influence the expression of genes associated with DAT endocytosis by modulating the chromatin's accessibility and compaction state. These enzymes control the expression of proteins involved in regulating endocytosis, promoting endosome formation, and facilitating recycling processes. Through the modulation exerted by these enzymes, the speed of DAT endocytosis and recycling patterns are indirectly regulated, establishing a crucial epigenetic control point for the regulation of neurotransmitter transport. Based on this understanding, we anticipate that targeting these processes could lead to favorable therapeutic effects for early PD pathogenesis.
Collapse
Affiliation(s)
- Ziqi Liang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201210, China
| | - Wanqing Liu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201210, China
| | - Mian Cao
- Program in Neuroscience and Behavioral Disorders, Duke-NUS Medical School, Singapore, 169857, Singapore; Department of Physiology, National University of Singapore, Singapore, 169857, Singapore
| | - Jiajun Cui
- Department of Biochemistry, College of Medicine, Yichun University, Yichun, Jiangxi 336000, China
| | - Jinshuai Lan
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201210, China
| | - Yue Ding
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201210, China
| | - Tong Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201210, China
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201210, China
| | - Zizhao Yang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201210, China
- Program in Neuroscience and Behavioral Disorders, Duke-NUS Medical School, Singapore, 169857, Singapore; Department of Physiology, National University of Singapore, Singapore, 169857, Singapore
- Department of General Surgery, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, 200137, China
| |
Collapse
|
25
|
Xu M, Liu X, Wang M, Luo T, Gao Y, Liu J, Shi J. BASAL: a universal mapping algorithm for nucleotide base-conversion sequencing. Nucleic Acids Res 2025; 53:gkae1201. [PMID: 39658059 PMCID: PMC11754667 DOI: 10.1093/nar/gkae1201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 11/13/2024] [Accepted: 11/19/2024] [Indexed: 12/12/2024] Open
Abstract
Utilizing base-conversion (BC) techniques, single-base resolution profiling of RNA and DNA modifications has significantly advanced. BC strategies range from one-way conversions (e.g. cytosine-to-thymine for 5-methylcytosine, adenine-to-guanine for N6-methyladenosine), to multi-way conversions (e.g. adenine to cytosine/guanine/thymine for N1-methyladenosine) and deletion-induced conversions (e.g. pseudouridine-to-deletion). Existing sequence aligners struggle with these diverse conversions, often leading to misaligning or inefficiency. We introduce BASAL (BAse-conversion Sequencing ALigner), which leverages bit-masking technology to accurately calculate mismatch penalties and supports all BC strategies. BASAL outperforms state-of-the-art tools in both mapping accuracy and efficiency. Through simulated and real data testing, along with experimental validation, we demonstrate that BASAL excels at identifying reliable modification sites. Moreover, BASAL enhances single-cell m6A analysis, revealing cell subpopulations and a cell evolutionary direction that align with biological functions, which other aligners fall short. BASAL's versatility establishes it as a universal aligner for RNA and DNA modification sequencing, facilitating groundbreaking discoveries in epigenomics and epitranscriptomics.
Collapse
Affiliation(s)
- Moping Xu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of the Ministry of Education, Orthopaedic Department of Tongji Hospital, Tongji University, 389 Xincun Road, Shanghai 200065, China
- Frontier Science Center for Stem Cell Research, Tongji University, 1239 Siping Road, Shanghai 200092, China
| | - Xiaoyang Liu
- State Key Laboratory of Protein and Plant Gene Research, Peking-Tsinghua Center for Life Sciences, School of Life Sciences, Peking University, 5 Yiheyuan Road, Beijing 100871, China
| | - Miao Wang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of the Ministry of Education, Orthopaedic Department of Tongji Hospital, Tongji University, 389 Xincun Road, Shanghai 200065, China
- Frontier Science Center for Stem Cell Research, Tongji University, 1239 Siping Road, Shanghai 200092, China
| | - Tingting Luo
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of the Ministry of Education, Orthopaedic Department of Tongji Hospital, Tongji University, 389 Xincun Road, Shanghai 200065, China
- Frontier Science Center for Stem Cell Research, Tongji University, 1239 Siping Road, Shanghai 200092, China
| | - Yawei Gao
- Department of Reproductive Medicine Center, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai 200092, China
| | - Jun Liu
- State Key Laboratory of Protein and Plant Gene Research, Peking-Tsinghua Center for Life Sciences, School of Life Sciences, Peking University, 5 Yiheyuan Road, Beijing 100871, China
- Beijing Advanced Center of RNA Biology (BEACON), Peking University, 5 Yiheyuan Road, Beijing 100871, China
| | - Jiejun Shi
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of the Ministry of Education, Orthopaedic Department of Tongji Hospital, Tongji University, 389 Xincun Road, Shanghai 200065, China
- Frontier Science Center for Stem Cell Research, Tongji University, 1239 Siping Road, Shanghai 200092, China
| |
Collapse
|
26
|
Chen F, Li X, Bai M, Zhao Y. Visualizing epigenetic modifications and their spatial proximities in single cells using three DNA-encoded amplifying FISH imaging strategies: BEA-FISH, PPDA-FISH and Cell-TALKING. Nat Protoc 2025; 20:220-247. [PMID: 39232201 DOI: 10.1038/s41596-024-01036-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 06/11/2024] [Indexed: 09/06/2024]
Abstract
Epigenetic modifications and spatial proximities of nucleic acids and proteins play important roles in regulating physiological processes and disease progression. Currently available cell imaging methods, such as fluorescence in situ hybridization (FISH) and immunofluorescence, struggle to detect low-abundance modifications and their spatial proximities. Here we describe a step-by-step protocol for three DNA-encoded amplifying FISH-based imaging strategies to overcome these challenges for varying applications: base-encoded amplifying FISH (BEA-FISH), pairwise proximity-differentiated amplifying FISH (PPDA-FISH) and cellular macromolecules-tethered DNA walking indexing (Cell-TALKING). They all use the similar core principle of DNA-encoded amplification, which transforms different nonsequence molecular features into unique DNA barcodes for in situ rolling circle amplification and FISH analysis. This involves three key reactions in fixed cell samples: target labeling, DNA encoding and rolling circle amplification imaging. Using this protocol, these three imaging strategies achieve in situ counting of low-abundance modifications alone, the pairwise proximity-differentiated visualization of two modifications and the exploration of multiple modifications around one protein (one-to-many proximity), respectively. Low-abundance modifications, including 5-hydroxymethylcytosine, 5-formylcytosine, 5-hydroxymethyluracil and 5-formyluracil, are clearly visualized in single cells. Various combinatorial patterns of nucleic acid modifications and/or histone modifications are found. The whole protocol takes ~2-4 d to complete, depending on different imaging applications.
Collapse
Affiliation(s)
- Feng Chen
- Institute of Analytical Chemistry and Instrument for Life Science, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'An, P. R. China
| | - Xinyin Li
- Institute of Analytical Chemistry and Instrument for Life Science, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'An, P. R. China
| | - Min Bai
- Institute of Analytical Chemistry and Instrument for Life Science, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'An, P. R. China
| | - Yongxi Zhao
- Institute of Analytical Chemistry and Instrument for Life Science, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'An, P. R. China.
| |
Collapse
|
27
|
Jiang J, Zhang Y, Wang J, Qin Y, Zhao C, He K, Wang C, Liu Y, Feng H, Cai H, He S, Li R, Galstyan DS, Yang L, Lim LW, de Abreu MS, Kalueff AV. Using Zebrafish Models to Study Epitranscriptomic Regulation of CNS Functions. J Neurochem 2025; 169:e16311. [PMID: 39825734 DOI: 10.1111/jnc.16311] [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: 10/11/2024] [Revised: 12/18/2024] [Accepted: 12/30/2024] [Indexed: 01/20/2025]
Abstract
Epitranscriptomic regulation of cell functions involves multiple post-transcriptional chemical modifications of coding and non-coding RNA that are increasingly recognized in studying human brain disorders. Although rodent models are presently widely used in neuroepitranscriptomic research, the zebrafish (Danio rerio) has emerged as a useful and promising alternative model species. Mounting evidence supports the importance of RNA modifications in zebrafish CNS function, providing additional insights into epitranscriptomic mechanisms underlying a wide range of brain disorders. Here, we discuss recent data on the role of RNA modifications in CNS regulation, with a particular focus on zebrafish models, as well as evaluate current problems, challenges, and future directions of research in this field of molecular neurochemistry.
Collapse
Affiliation(s)
- Jiayou Jiang
- Suzhou Municipal Key Laboratory of Neurobiology and Cell Signaling, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China
- Department of Biosciences and Bioinformatics, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China
| | - Yunqian Zhang
- Suzhou Municipal Key Laboratory of Neurobiology and Cell Signaling, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China
- Department of Biosciences and Bioinformatics, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China
| | - Jiyi Wang
- Suzhou Municipal Key Laboratory of Neurobiology and Cell Signaling, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China
- Department of Biosciences and Bioinformatics, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China
| | - Yixin Qin
- Suzhou Municipal Key Laboratory of Neurobiology and Cell Signaling, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China
- Department of Biosciences and Bioinformatics, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China
| | - Chonguang Zhao
- Suzhou Municipal Key Laboratory of Neurobiology and Cell Signaling, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China
- Department of Biosciences and Bioinformatics, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China
| | - Kai He
- Suzhou Municipal Key Laboratory of Neurobiology and Cell Signaling, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China
- Department of Biosciences and Bioinformatics, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China
| | - Chaoming Wang
- Suzhou Municipal Key Laboratory of Neurobiology and Cell Signaling, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China
- Department of Biosciences and Bioinformatics, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China
| | - Yucheng Liu
- Suzhou Municipal Key Laboratory of Neurobiology and Cell Signaling, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China
- Department of Biosciences and Bioinformatics, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China
| | - Haoyu Feng
- Suzhou Municipal Key Laboratory of Neurobiology and Cell Signaling, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China
- Department of Biosciences and Bioinformatics, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China
| | - Huiling Cai
- Suzhou Municipal Key Laboratory of Neurobiology and Cell Signaling, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China
- Department of Biosciences and Bioinformatics, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China
| | - Shulei He
- Suzhou Municipal Key Laboratory of Neurobiology and Cell Signaling, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China
- Department of Biosciences and Bioinformatics, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China
| | - Ruiyu Li
- Suzhou Municipal Key Laboratory of Neurobiology and Cell Signaling, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China
- Department of Biosciences and Bioinformatics, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China
| | - David S Galstyan
- Institute of Experimental Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - Longen Yang
- Suzhou Municipal Key Laboratory of Neurobiology and Cell Signaling, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China
- Department of Biosciences and Bioinformatics, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China
| | - Lee Wei Lim
- Suzhou Municipal Key Laboratory of Neurobiology and Cell Signaling, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China
- Department of Biosciences and Bioinformatics, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China
| | - Murilo S de Abreu
- Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre, Porto Alegre, Brazil
- Moscow Institute of Physics and Technology, Moscow, Russia
- Western Caspian University, Baku, Azerbaijan
| | - Allan V Kalueff
- Suzhou Municipal Key Laboratory of Neurobiology and Cell Signaling, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China
- Department of Biosciences and Bioinformatics, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China
- Moscow Institute of Physics and Technology, Moscow, Russia
| |
Collapse
|
28
|
Chen X, Xu H, Shu X, Song CX. Mapping epigenetic modifications by sequencing technologies. Cell Death Differ 2025; 32:56-65. [PMID: 37658169 PMCID: PMC11742697 DOI: 10.1038/s41418-023-01213-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 08/09/2023] [Accepted: 08/14/2023] [Indexed: 09/03/2023] Open
Abstract
The "epigenetics" concept was first described in 1942. Thus far, chemical modifications on histones, DNA, and RNA have emerged as three important building blocks of epigenetic modifications. Many epigenetic modifications have been intensively studied and found to be involved in most essential biological processes as well as human diseases, including cancer. Precisely and quantitatively mapping over 100 [1], 17 [2], and 160 [3] different known types of epigenetic modifications in histone, DNA, and RNA is the key to understanding the role of epigenetic modifications in gene regulation in diverse biological processes. With the rapid development of sequencing technologies, scientists are able to detect specific epigenetic modifications with various quantitative, high-resolution, whole-genome/transcriptome approaches. Here, we summarize recent advances in epigenetic modification sequencing technologies, focusing on major histone, DNA, and RNA modifications in mammalian cells.
Collapse
Affiliation(s)
- Xiufei Chen
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7FZ, UK
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7FZ, UK
| | - Haiqi Xu
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7FZ, UK
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7FZ, UK
| | - Xiao Shu
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7FZ, UK
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7FZ, UK
| | - Chun-Xiao Song
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7FZ, UK.
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7FZ, UK.
| |
Collapse
|
29
|
ZHANG L, ZHANG W, WANG H. [Current advances in the analysis of free RNA modified nucleosides by high performance liquid chromatography-tandem mass spectrometry]. Se Pu 2025; 43:3-12. [PMID: 39722616 PMCID: PMC11686471 DOI: 10.3724/sp.j.1123.2024.07004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Indexed: 12/28/2024] Open
Abstract
Post-transcriptional ribonucleic acid (RNA) modifications play crucial roles in regulating gene expression, with both eukaryotic and prokaryotic RNA exhibiting more than 170 distinct and ubiquitous modifications. RNA turnover generates numerous free nucleosides, including unmodified nucleosides and a variety of modified ones. Unlike unmodified nucleosides, modified nucleosides are not further degraded or used in the salvage-synthesis pathway owing to a lack of specific enzymes, which leads to the cytosolic accumulation or cellular efflux of modified nucleosides. These modified nucleosides can act as signaling molecules that regulate downstream pathways once transported to the extracellular space; alternatively, they are metabolized in the bloodstream and excreted in urine. Metabolized modified nucleosides are altered by cellular stress responses and mediate abnormal physiological states. Changes in the urinary and blood levels of modified nucleosides associated with cancer can serve as biomarkers for disease. Therefore, identifying and accurately quantifying nucleosides is vital for understanding RNA degradation and associated patterns of nucleoside metabolism. Such analyses are helpful when studying the biological functions and potential clinical applications of modified nucleosides. In this regard, high performance liquid chromatography-tandem mass spectrometry (HPLC-MS/MS) offers significant advantages in terms of sensitivity, selectivity, and efficiency, and has been widely used to analyze DNA and RNA nucleosides/nucleotides and their analogues. Multiple MS detection patterns and quantification methods have been established to detect nucleosides in biological samples, including cultured cells, urine, blood, and tissue samples. However, the development of an accurate HPLC-MS/MS method faces several challenges. Firstly, the presence of a complex biological matrix that contains macromolecules, small molecules, and salts can interfere with analysis. Salts and co-eluting substances in the extraction solution often affect mass-spectrometric responses for target analytes. Secondly, various nucleosides are present in vastly different abundances, with contents varying by up to four orders of magnitude; hence, accurately quantifying multiple nucleosides in a single assay is challenging. Thirdly, N-glycosidic bonds are favorably cleaved in most nucleosides during MS to produce the same characteristic fragment ions, which are often accompanied by nucleobases. This tendency poses challenges for distinguishing structural isomers and mass-analogs of modified nucleosides by MS. Post-transcriptional chemical modifications include methylation, hydroxylation, sulfur/oxygen substitution, and side-chain additions. Developing a unified method for simultaneously screening modified nucleosides is difficult owing to biochemical diversity; consequently, there is a need for advanced HPLC-MS/MS method capable of accurately quantifying such nucleosides. This review summarizes the development and applications of LC-MS technologies for analyzing endogenous nucleosides, covering sample preparation, chromatographic-separation and mass-spectrometric-detection conditions, and the development of quantification methods. Additionally, we discuss applications aimed at detecting and quantifying RNA-derived modified nucleosides in biological samples. The applications of HPLC-MS/MS technology are highlighted, the regulation and function of free modified nucleosides are discussed, and the potential functions of modified nucleosides as disease biomarkers for clinical applications are introduced.
Collapse
|
30
|
Meng J, Yan C, Liu J. LDHA- Mediated Histone Lactylation Promotes the Nonalcoholic Fatty Liver Disease Progression Through Targeting The METTL3/ YTHDF1/SCD1 m6A Axis. Physiol Res 2024; 73:985-999. [PMID: 39903889 PMCID: PMC11835221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 08/01/2024] [Indexed: 02/06/2025] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is characterized by elevated hepatic lipids caused by nonalcoholic factors, where histone lactylation is lately discovered as a modification driving disease progression. This research aimed to explore the role of histone 3 lysine 18 lactylation (H3K18lac) in NAFLD progression using a high-fat diet (HFD)-treated mouse model and free fatty acids (FFA)-treated L-02 cell lines. Lipids accumulation was screened via Oil Red O staining, real-time quantitative polymerase chain reaction (RT-qPCR), western blotting, and commercially available kits. Similarly, molecular mechanism was analyzed using immunoprecipitation (IP), dual-luciferase reporter assay, and RNA decay assay. Results indicated that FFA upregulated lactate dehydrogenase A (LDHA) and H3K18lac levels in L-02 cells. Besides, LDHA-mediated H3K18lac was enriched on the proximal promoter of methyltransferase 3 (METTL3), translating into an increased expression. Moreover, METTL3 or LDHA knockdown relieved lipid accumulation, decreased total cholesterol (TC) and triglyceride (TG) levels, and downregulated lipogenesis-related proteins in FFA-treated L-02 cell lines, in addition to enhancing the m6A and mRNA levels of stearoyl-coenzyme A desaturase 1 (SCD1). The m6A modification of SCD1 was recognized by YTH N6-methyladenosine RNA binding protein F1 (YTHDF1), resulting in enhanced mRNA stability. LDHA was found to be highly expressed in HFD-treated mice, where knocking down LDHA attenuated HFD-induced hepatic steatosis. These findings demonstrated that LDHA-induced H3K18lac promoted NAFLD progression, where LDHA-induced H3K18lac in METTL3 promoter elevated METTL3 expression, thereby promoting m6A methylation and stabilizing SCD1 via a YTHDF1-dependent manner. Keywords: Nonalcoholic fatty liver disease, LDHA, METTL3, YTHDF1, Histone lactylation.
Collapse
Affiliation(s)
- J Meng
- Shanxi Medical University, Yingze District, Taiyuan, Shanxi, China
| | | | | |
Collapse
|
31
|
Zhu H, Yang Y, Zhou Z. Construction and clinical application of a risk model based on N6-methyladenosine regulators for colorectal cancer. PeerJ 2024; 12:e18719. [PMID: 39717046 PMCID: PMC11665428 DOI: 10.7717/peerj.18719] [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: 09/19/2024] [Accepted: 11/26/2024] [Indexed: 12/25/2024] Open
Abstract
Background Colorectal cancer (CRC) shows a high incidence in developed countries. This study established a prognosis signature based on N6-methyladenosine (m6A) regulators involved in CRC progression. Method The bulk RNA-seq data from the Atlas and Compass of Immune-Colon cancer-Microbiome interactions (AC-ICAM) and GSE33113 CRC datasets were obtained from the cBioportal and GEO databases, and a total of 21 m6A regulators genes were collected from a previous study. The scRNA-seq analysis of the GSE146771 cohort was conducted applying the Seurat and harmony R packages. Consensus clustering based on the expressions of m6A regulators was performed with the ConsensusClusterPlus package. The ggGSEA package was used for the Gene Set Enrichment Analysis (GSEA). The un/multivariate and LASSO Cox analysis were performed applying the "survival" and "glmnet" packages for developing a risk model. The pRRophetic and GSVA packages were utilized to analyze potential drugs for CRC and immune infiltration in different risk groups, followed by the Kaplan-Meier (KM) survival and ROC analysis with the "survival" and "timeROC" packages. In vitro assays included the quantitative polymerase chain reaction (qPCR), wound healing and transwell were performed. Results CRC patients in the AC-ICAM cohort were assigned into three molecular subtypes (S1, 2 and 3) based on nine m6A regulator genes. Specifically, the prognostic outcome of the S3 was the most favorable, while that of the S1 was the worst and this subtype was associated with the activation of NF-kB, TNF-α and hypoxia pathways. Three key genes, namely, methyltransferase-like 3 (METTL3), insulinolike Growth Factor2 mRNA-Binding Protein 3 (IGF2BP3) and YTH domain-containing protein 2 (YTHDC2), selected from the 9 m6A regulator genes were combined into a RiskScore, which showed a high classification effectiveness in dividing the patients into high- and low-risk groups. Inhibition of the expression of METTL3A or that of IGF2BP3 suppressed the invasion and migration of CRC cells. Notably, the high-risk patients had higher immune cell infiltration to support the activation of multiple immune responses and exhibited significantly poor prognosis. Meanwhile, a nomogram with practical clinical value was developed based on the RiskScore and other clinical features. Finally, eight potential drugs associated with the RiskScore were identified, and CD4+ cells and Tregs were found to be closely associated with CRC progression. Conclusion The RiskScore model developed based on m6A regulators played a critical role in CRC development and can be considered as a prognosis predictor for patients with the cancer. The present discoveries will facilitate the diagnosis and clinical management of CRC patients.
Collapse
Affiliation(s)
- Hanhan Zhu
- Oncology Department, The Sixth Affiliated Hospital of Jinan University, Dongguan, China
| | - Yu Yang
- Oncology Department, The Sixth Affiliated Hospital of Jinan University, Dongguan, China
| | - Zhenfeng Zhou
- Cancer Diagnosis and Treatment Research Center, The First Affiliated Hospital of Jinan University, Guangzhou, China
| |
Collapse
|
32
|
Feng Q, Yu L, Li L, Zhang Q. Covalent inhibitors meet epigenetics: New opportunities. Eur J Med Chem 2024; 280:116951. [PMID: 39406112 DOI: 10.1016/j.ejmech.2024.116951] [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: 06/20/2024] [Revised: 08/09/2024] [Accepted: 09/23/2024] [Indexed: 11/25/2024]
Abstract
Epigenetic intervention has become an important therapeutic strategy for a variety of diseases, such as cancer. Although a small number of epigenetic drugs have been marketed, most of these inhibitors are limited by their poor efficacy, dose-dependent toxicity, poor selectivity, and drug resistance. The development of covalent inhibitors has progressed from questioning to resurgence. Its slow dissociation is expected to inject new vitality into epigenetic drugs. In this review, more than 40 covalent inhibitors of 29 epigenetic targets were collated, focusing on their design strategies, reaction mechanisms, covalent warheads and targeted amino acids, and covalent verification methods. Furthermore, this review presented new opportunities based on the current development of covalent inhibitors targeting epigenetic regulators. It is believed that epigenetic covalent inhibitors will lead to more breakthroughs.
Collapse
Affiliation(s)
- Qiang Feng
- College of Chemistry and Life Science, Sichuan Provincial Key Laboratory for Structural Optimization and Application of Functional Molecules, Chengdu Normal University, Chengdu, 611130, China
| | - Luoting Yu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, And Collaborative Innovation Center for Biotherapy, 17#3rd Section, Ren Min South Road, Chengdu 610041, China
| | - Lu Li
- Department of Pharmacy, NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Clinical Trial Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Qiangsheng Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, And Collaborative Innovation Center for Biotherapy, 17#3rd Section, Ren Min South Road, Chengdu 610041, China; Department of Pharmacy, West China Second University Hospital, Sichuan University, Children's Medicine Key Laboratory of Sichuan Province, Chengdu, 610041, China.
| |
Collapse
|
33
|
Han Y, Dong L, Zhu LY, Hu CR, Li H, Zhang Y, Zhang C, Zhang Y, Dong ZC. Real-Space Spectral Determination of Short Single-Stranded DNA Sequence Structures. J Am Chem Soc 2024; 146:33865-33873. [PMID: 39604067 DOI: 10.1021/jacs.4c12393] [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: 11/29/2024]
Abstract
Resolving the sequence and structure of flexible biomolecules such as DNA is crucial to understanding their biological mechanisms and functions. Traditional structural biology methods remain challenging for the analysis of small and disordered biomolecules, especially those that are difficult to label or crystallize. Recent development of single-molecule tip-enhanced Raman spectroscopy (TERS) offers a label-free approach to identifying nucleobases in a single DNA chain. However, a clear demonstration of sequencing both spatially and spectrally at single-base resolution is still elusive due to the challenges caused by weak Raman signals and the flexibility of DNA molecules. Here, we report a proof-of-principle demonstration to this end, spectrally resolving in real space individual nucleobases and their sequence structures within a short, single-stranded DNA molecule artificially designed. This breakthrough is achieved through the development of subnanometer-resolved low-temperature TERS methodology for such thermally unstable flexible biomolecules. Further TERS mapping over individual nucleobases provides additional structural information about the molecular configurations and even the locations of functional groups, offering a way to track modification types and binding sites in biomolecules.
Collapse
Affiliation(s)
- Yu Han
- Hefei National Research Center for Physical Sciences at the Microscale and CAS Center for Excellence in Quantum Information and Quantum Physics, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Li Dong
- Hefei National Research Center for Physical Sciences at the Microscale and CAS Center for Excellence in Quantum Information and Quantum Physics, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Lu-Yao Zhu
- Hefei National Research Center for Physical Sciences at the Microscale and CAS Center for Excellence in Quantum Information and Quantum Physics, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Chun-Rui Hu
- Hefei National Research Center for Physical Sciences at the Microscale and CAS Center for Excellence in Quantum Information and Quantum Physics, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Hang Li
- Hefei National Research Center for Physical Sciences at the Microscale and CAS Center for Excellence in Quantum Information and Quantum Physics, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Yang Zhang
- Hefei National Research Center for Physical Sciences at the Microscale and CAS Center for Excellence in Quantum Information and Quantum Physics, University of Science and Technology of China, Hefei, Anhui 230026, China
- School of Physics and Department of Chemical Physics, University of Science and Technology of China, Hefei, Anhui 230026, China
- Hefei National Laboratory, University of Science and Technology of China, Hefei, Anhui 230088, China
- Key Laboratory of Precision and Intelligent Chemistry, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Chao Zhang
- Hefei National Research Center for Physical Sciences at the Microscale and CAS Center for Excellence in Quantum Information and Quantum Physics, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Yao Zhang
- Hefei National Research Center for Physical Sciences at the Microscale and CAS Center for Excellence in Quantum Information and Quantum Physics, University of Science and Technology of China, Hefei, Anhui 230026, China
- School of Physics and Department of Chemical Physics, University of Science and Technology of China, Hefei, Anhui 230026, China
- Hefei National Laboratory, University of Science and Technology of China, Hefei, Anhui 230088, China
| | - Zhen-Chao Dong
- Hefei National Research Center for Physical Sciences at the Microscale and CAS Center for Excellence in Quantum Information and Quantum Physics, University of Science and Technology of China, Hefei, Anhui 230026, China
- School of Physics and Department of Chemical Physics, University of Science and Technology of China, Hefei, Anhui 230026, China
- Hefei National Laboratory, University of Science and Technology of China, Hefei, Anhui 230088, China
| |
Collapse
|
34
|
Li Y, Guo X, Yao H, Zhang Z, Zhao H. Epigenetic control of dental stem cells: progress and prospects in multidirectional differentiation. Epigenetics Chromatin 2024; 17:37. [PMID: 39623487 PMCID: PMC11613947 DOI: 10.1186/s13072-024-00563-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 11/26/2024] [Indexed: 12/06/2024] Open
Abstract
Dental stem cells, with their exceptional proliferative capacity and multidirectional differentiation potential, hold significant promise for dental and oral tissue regeneration. Epigenetic inheritance, which involves stable and heritable changes in gene expression and function without alterations to the DNA sequence, plays a critical role in numerous biological processes. Environmental factors are particularly influential in epigenetic inheritance, as variations in exposure can lead to changes in epigenetic modifications that subsequently impact gene expression. Epigenetic mechanisms are widely involved in processes such as bone homeostasis, embryogenesis, stem cell fate determination, and disease development. Recently, the epigenetic regulation of dental stem cells has attracted considerable research attention. This paper reviews studies focused on the epigenetic mechanisms governing the multidirectional differentiation of dental stem cells.
Collapse
Affiliation(s)
- Yan Li
- Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Xinwei Guo
- Department of Stomatology, The First Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Hua Yao
- Department of Stomatology, The First Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Zhimin Zhang
- Hospital of Stomatology, Jilin University, Changchun, 130021, China.
| | - Hongyan Zhao
- Hospital of Stomatology, Jilin University, Changchun, 130021, China.
| |
Collapse
|
35
|
Wang X, Ma X, Chen S, Fan M, Jin C, Chen Y, Wang S, Wang Z, Meng F, Zhang C, Yang L. Harnessing m1A modification: a new frontier in cancer immunotherapy. Front Immunol 2024; 15:1517604. [PMID: 39687616 PMCID: PMC11647001 DOI: 10.3389/fimmu.2024.1517604] [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: 10/26/2024] [Accepted: 11/18/2024] [Indexed: 12/18/2024] Open
Abstract
N1-methyladenosine (m1A) modification is an epigenetic change that occurs on RNA molecules, regulated by a suite of enzymes including methyltransferases (writers), demethylases (erasers), and m1A-recognizing proteins (readers). This modification significantly impacts the function of RNA and various biological processes by affecting the structure, stability, translation, metabolism, and gene expression of RNA. Thereby, m1A modification is closely associated with the occurrence and progression of cancer. This review aims to explore the role of m1A modification in tumor immunity. m1A affects tumor immune responses by directly regulating immune cells and indirectly modulating tumor microenvironment. Besides, we also discuss the implications of m1A-mediated metabolic reprogramming and its nexus with immune checkpoint inhibitors, unveiling promising avenues for immunotherapeutic intervention. Additionally, the m1AScore, established based on the expression patterns of m1A modification, can be used to predict tumor prognosis and guide personalized therapy. Our review underscores the significance of m1A modification as a burgeoning frontier in cancer biology and immuno-oncology, with the potential to revolutionize cancer treatment strategies.
Collapse
Affiliation(s)
- Xinru Wang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Xiaoqing Ma
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Siyu Chen
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Minyan Fan
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Chenying Jin
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Yushi Chen
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Shaodong Wang
- Affiliated Nanjing Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, China
| | - Zhiying Wang
- Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao, Shandong, China
| | - Fei Meng
- Department of Clinical Laboratory, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Chengwan Zhang
- Department of Central Laboratory, The Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical University, Huai’an, Jiangsu, China
| | - Lin Yang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| |
Collapse
|
36
|
Cui X, Li H, Huang X, Xue T, Wang S, Zhu X, Jing X. N 6-Methyladenosine Modification on the Function of Female Reproductive Development and Related Diseases. Immun Inflamm Dis 2024; 12:e70089. [PMID: 39660878 PMCID: PMC11632877 DOI: 10.1002/iid3.70089] [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: 08/18/2024] [Revised: 10/17/2024] [Accepted: 11/20/2024] [Indexed: 12/12/2024] Open
Abstract
BACKGROUND N6-methyladenosine (m6A) modification is a widespread and reversible epigenetic alteration in eukaryotic mRNA, playing a pivotal role in various biological functions. Its significance in female reproductive development and associated diseases has recently become a focal point of research. OBJECTIVE This review aims to consolidate current knowledge of the role of m6A modification in female reproductive tissues, emphasizing its regulatory dynamics, functional significance, and implications in reproductive health and disease. METHODS A comprehensive analysis of recent studies focusing on m6A modification in ovarian development, oocyte maturation, embryo development, and the pathogenesis of reproductive diseases. RESULTS m6A modification exhibits dynamic regulation in female reproductive tissues, influencing key developmental stages and processes. It plays critical roles in ovarian development, oocyte maturation, and embryo development, underpinning essential aspects of reproductive health. m6A modification is intricately involved in the pathogenesis of several reproductive diseases, including polycystic ovary syndrome (PCOS), premature ovarian failure (POF), and endometriosis, offering insights into potential molecular mechanisms and therapeutic targets. CONCLUSION The review highlights the crucial role of m6A modification in female reproductive development and related diseases. It underscores the need for further research to explore innovative diagnostic and therapeutic strategies for reproductive disorders, leveraging the insights gained from understanding m6A modification's impact on reproductive health.
Collapse
Affiliation(s)
- Xiangrong Cui
- Reproductive Medicine CenterThe affiliated Children's Hospital of Shanxi Medical University, Children's Hospital of Shanxi, Shanxi Maternal and Child Health HospitalTaiyuanChina
| | - Huihui Li
- Reproductive Medicine CenterThe affiliated Children's Hospital of Shanxi Medical University, Children's Hospital of Shanxi, Shanxi Maternal and Child Health HospitalTaiyuanChina
| | - Xia Huang
- Department of Clinical LaboratoryShanxi Provincial People's Hospital, Shanxi Medical UniversityTaiyuanChina
| | - Tingting Xue
- Department of Clinical LaboratoryShanxi Provincial People's Hospital, Shanxi Medical UniversityTaiyuanChina
| | - Shu Wang
- Department of Clinical LaboratoryShanxi Provincial People's Hospital, Shanxi Medical UniversityTaiyuanChina
| | - Xinyu Zhu
- Department of Clinical LaboratoryShanxi Provincial People's Hospital, Shanxi Medical UniversityTaiyuanChina
| | - Xuan Jing
- Department of Clinical LaboratoryShanxi Provincial People's Hospital, Shanxi Medical UniversityTaiyuanChina
| |
Collapse
|
37
|
Fiordoro S, Rosano C, Pechkova E, Barocci S, Izzotti A. Epigenetic modulation of immune cells: Mechanisms and implications. Adv Biol Regul 2024; 94:101043. [PMID: 39305736 DOI: 10.1016/j.jbior.2024.101043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 07/25/2024] [Accepted: 08/02/2024] [Indexed: 12/12/2024]
Abstract
Epigenetic modulation of the immune response entails modifiable and inheritable modifications that do not modify the DNA sequence. While there have been many studies on epigenetic changes in tumor cells, there is now a growing focus on epigenetically mediated changes in immune cells of both the innate and adaptive systems. These changes have significant implications for both the body's response to tumors and the development of potential therapeutic vaccines. This study primarily discusses the key epigenetic alterations, with a specific emphasis on pseudouridination, as well as non-coding RNAs and their transportation, which can lead to the development of cancer and the acquisition of new phenotypic traits by immune cells. Furthermore, the advancement of therapeutic vaccinations targeting the tumor will be outlined.
Collapse
Affiliation(s)
- S Fiordoro
- Department of Health Sciences, University of Genova, Via Pastore 1, 16132 Genova, Italy
| | - C Rosano
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy.
| | - E Pechkova
- Department of Experimental Medicine, University of Genoa, 16132 Genova, Italy
| | - S Barocci
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy
| | - A Izzotti
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy; Department of Experimental Medicine, University of Genoa, 16132 Genova, Italy
| |
Collapse
|
38
|
Zhao Z, Yang T, Li F. Sperm RNA code in spermatogenesis and male infertility. Reprod Biomed Online 2024; 49:104375. [PMID: 39481211 DOI: 10.1016/j.rbmo.2024.104375] [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: 02/27/2024] [Revised: 05/22/2024] [Accepted: 07/05/2024] [Indexed: 11/02/2024]
Abstract
Spermatozoa are traditionally thought to be transcriptionally inert, but recent studies have revealed the presence of sperm RNA, some of which is derived from the residues of spermatocyte transcription and some from epididymosomes. Paternal sperm RNA can be affected by external factors and further modified at the post-transcriptional level, for example N6-methyladenosine (m6A), thus shaping spermatogenesis and reproductive outcome. This review briefly introduces the origin of sperm RNA and, on this basis, summarizes the current knowledge on RNA modifications and their functional role in spermatogenesis and male infertility. The bottlenecks and knowledge gaps in the current research on RNA modification in male reproduction have also been indicated. Further investigations are needed to elucidate the functional consequences of these modifications, providing new therapeutic and preventive strategies for reproductive health and genetic inheritance.
Collapse
Affiliation(s)
- Zhongyi Zhao
- Department of Andrology/Sichuan Human Sperm Bank, West China Second University Hospital, Sichuan University, Chengdu, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Tingting Yang
- Department of Andrology/Sichuan Human Sperm Bank, West China Second University Hospital, Sichuan University, Chengdu, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China.
| | - Fuping Li
- Department of Andrology/Sichuan Human Sperm Bank, West China Second University Hospital, Sichuan University, Chengdu, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China.
| |
Collapse
|
39
|
Belizário J, Garay-Malpartida M. Key Epigenetic Players in Etiology and Novel Combinatorial Therapies for Treatment of Hepatocellular Carcinoma. LIVERS 2024; 4:638-655. [DOI: 10.3390/livers4040044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the leading causes of death in which the molecular tumorigenesis and cellular heterogeneity are poorly understood. The genetic principle that specific driver mutations in oncogenes, DNA repair genes, and tumor-suppressor genes can independently drive cancer development has been widely explored. Additionally, a repertory of harmful epigenetic modifications in DNA and chromatin—impacting the expression of genes involved in cellular proliferation, differentiation, genome stability, cell-cycle control, and DNA repair—are now acknowledged across various biological contexts that contribute to cancer etiology. Notably, the dynamic hypermethylation and hypomethylation in enhancer and promoter regions that promote activation or silencing of the master regulatory genes of the epigenetic programs is often altered in tumor cells due to mutation. Genome instability is one of the cancer hallmarks that contribute to transdifferentiation and intratumoral heterogeneity. Thus, it is broadly accepted that tumor tissue is dominated by genetically and epigenetically distinct sub-clones which display a set of genetic and epigenetic mutations. Here we summarize some functions of key genetic and epigenetic players and biochemical pathways leading to liver cell transformation. We discuss the role of the potential epigenetic marks in target genes thought to be involved in sequential events following liver lipid metabolism dysregulation, inflammation, fibrosis, cirrhosis, and finally hepatocellular carcinoma. We also briefly describe new findings showing how epigenetic drugs together with chemotherapy and immunotherapy can improve overall responses in patients with hepatic tumors.
Collapse
Affiliation(s)
- José Belizário
- School of Arts, Sciences and Humanities of the University of Sao Paulo, Rua Arlindo Bettio, 1000, São Paulo 03828-000, Brazil
| | - Miguel Garay-Malpartida
- School of Arts, Sciences and Humanities of the University of Sao Paulo, Rua Arlindo Bettio, 1000, São Paulo 03828-000, Brazil
| |
Collapse
|
40
|
Henkel M, Fillbrunn A, Marchand V, Raghunathan G, Berthold MR, Motorin Y, Marx A. A DNA Polymerase Variant Senses the Epigenetic Marker 5-Methylcytosine by Increased Misincorporation. Angew Chem Int Ed Engl 2024; 63:e202413304. [PMID: 39449390 DOI: 10.1002/anie.202413304] [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: 07/15/2024] [Indexed: 10/26/2024]
Abstract
Dysregulation of DNA methylation is associated with human disease, particularly cancer, and the assessment of aberrant methylation patterns holds great promise for clinical diagnostics. However, DNA polymerases do not effectively discriminate between processing 5-methylcytosine (5 mC) and unmethylated cytosine, resulting in the silencing of methylation information during amplification or sequencing. As a result, current detection methods require multi-step DNA conversion treatments or careful analysis of sequencing data to decipher individual 5 mC bases. To overcome these challenges, we propose a novel DNA polymerase-mediated 5 mC detection approach. Here, we describe the engineering of a thermostable DNA polymerase variant derived from Thermus aquaticus with altered fidelity towards 5 mC. Using a screening-based evolutionary approach, we have identified a DNA polymerase that exhibits increased misincorporation towards 5 mC during DNA synthesis. This DNA polymerase generates mutation signatures at methylated CpG sites, allowing direct detection of 5 mC by reading an increased error rate after sequencing without prior treatment of the sample DNA.
Collapse
Affiliation(s)
- Melanie Henkel
- Department of Chemistry, Konstanz Research School Chemical Biology, University of Konstanz, Universitätsstraße 10, 78464, Konstanz, Germany
| | - Alexander Fillbrunn
- Department of Computer Science, Konstanz Research School Chemical Biology, University of Konstanz, Universitätsstraße 10, 78464, Konstanz, Germany
| | - Virginie Marchand
- Epitranscriptomics and Sequencing (EpiRNA-Seq) Core Facility, UAR2008/US40 Ingénierie Biologie Santé en Lorraine (IBSLor), CNRS-UL-INSERM, Université de Lorraine, 9 Avenue de la Forêt de Haye, BP 20199, 54505, Vandoeuvre-les-Nancy, France
| | - Govindan Raghunathan
- Department of Chemistry, Konstanz Research School Chemical Biology, University of Konstanz, Universitätsstraße 10, 78464, Konstanz, Germany
| | - Michael R Berthold
- Department of Computer Science, Konstanz Research School Chemical Biology, University of Konstanz, Universitätsstraße 10, 78464, Konstanz, Germany
- KNIME AG, Talacker 50, 8001, Zurich, Switzerland
| | - Yuri Motorin
- Epitranscriptomics and Sequencing (EpiRNA-Seq) Core Facility, UAR2008/US40 Ingénierie Biologie Santé en Lorraine (IBSLor), CNRS-UL-INSERM, Université de Lorraine, 9 Avenue de la Forêt de Haye, BP 20199, 54505, Vandoeuvre-les-Nancy, France
- Ingénierie Moléculaire et Physiopathologie Articulaire (IMoPA), UMR7365 CNRS-Université de Lorraine, Université de Lorraine, 9 Avenue de la Forêt de Haye, BP 20199, 54505, Vandoeuvre-les-Nancy, France
| | - Andreas Marx
- Department of Chemistry, Konstanz Research School Chemical Biology, University of Konstanz, Universitätsstraße 10, 78464, Konstanz, Germany
| |
Collapse
|
41
|
Wang H, Tang J, Yan S, Li C, Li Z, Xiong Z, Li Z, Tu C. Liquid-liquid Phase Separation in Aging: Novel Insights in the Pathogenesis and Therapeutics. Ageing Res Rev 2024; 102:102583. [PMID: 39566743 DOI: 10.1016/j.arr.2024.102583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 10/14/2024] [Accepted: 11/12/2024] [Indexed: 11/22/2024]
Abstract
The intricate organization of distinct cellular compartments is paramount for the maintenance of normal biological functions and the orchestration of complex biochemical reactions. These compartments, whether membrane-bound organelles or membraneless structures like Cajal bodies and RNA transport granules, play crucial roles in cellular function. Liquid-liquid phase separation (LLPS) serves as a reversible process that elucidates the genesis of membranelles structures through the self-assembly of biomolecules. LLPS has been implicated in a myriad of physiological and pathological processes, encompassing immune response and tumor genesis. But the association between LLPS and aging has not been clearly clarified. A recent advancement in the realm of aging research involves the introduction of a new edition outlining the twelve hallmarks of aging, categorized into three distinct groups. By delving into the role and mechanism of LLPS in the formation of membraneless structures at a molecular level, this review encapsulates an exploration of the interaction between LLPS and these aging hallmarks, aiming to offer novel perspectives of the intricate mechanisms underlying the aging process and deeper insights into aging therapeutics.
Collapse
Affiliation(s)
- Hua Wang
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University Changsha 410011, China
| | - Jinxin Tang
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University Changsha 410011, China
| | - Shuxiang Yan
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, Institute of Nephrology, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Chenbei Li
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University Changsha 410011, China
| | - Zhaoqi Li
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University Changsha 410011, China
| | - Zijian Xiong
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University Changsha 410011, China
| | - Zhihong Li
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University Changsha 410011, China; Hunan Key Laboratory of Tumor Models and Individualized Medicine, Engineering Research Center of Artificial Intelligence-Driven Medical Device, The Second Xiangya Hospital of Central South University Changsha 410011, China, Changsha 410011, China; Shenzhen Research Institute of Central South University, Shenzhen 518063, China
| | - Chao Tu
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University Changsha 410011, China; Changsha Medical University, Changsha 410219, China
| |
Collapse
|
42
|
Mu S, Wang W, Liu Q, Ke N, Li H, Sun F, Zhang J, Zhu Z. Autoimmune disease: a view of epigenetics and therapeutic targeting. Front Immunol 2024; 15:1482728. [PMID: 39606248 PMCID: PMC11599216 DOI: 10.3389/fimmu.2024.1482728] [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: 08/23/2024] [Accepted: 10/23/2024] [Indexed: 11/29/2024] Open
Abstract
Autoimmune diseases comprise a large group of conditions characterized by a complex pathogenesis and significant heterogeneity in their clinical manifestations. Advances in sequencing technology have revealed that in addition to genetic susceptibility, various epigenetic mechanisms including DNA methylation and histone modification play critical roles in disease development. The emerging field of epigenetics has provided new perspectives on the pathogenesis and development of autoimmune diseases. Aberrant epigenetic modifications can be used as biomarkers for disease diagnosis and prognosis. Exploration of human epigenetic profiles revealed that patients with autoimmune diseases exhibit markedly altered DNA methylation profiles compared with healthy individuals. Targeted cutting-edge epigenetic therapies are emerging. For example, DNA methylation inhibitors can rectify methylation dysregulation and relieve patients. Histone deacetylase inhibitors such as vorinostat can affect chromatin accessibility and further regulate gene expression, and have been used in treating hematological malignancies. Epigenetic therapies have opened new avenues for the precise treatment of autoimmune diseases and offer new opportunities for improved therapeutic outcomes. Our review can aid in comprehensively elucidation of the mechanisms of autoimmune diseases and development of new targeted therapies that ultimately benefit patients with these conditions.
Collapse
Affiliation(s)
- Siqi Mu
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, Anhui, China
- Department of Skin Genetics, Anhui Province Laboratory of Inflammation and Immune Mediated Diseases, Hefei, Anhui, China
- Department of Dermatology, Shannan People's Hospital, Shannan, China
- First Clinical Medical College, Anhui Medical University, Hefei, Anhui, China
| | - Wanrong Wang
- Department of Skin Genetics, Anhui Province Laboratory of Inflammation and Immune Mediated Diseases, Hefei, Anhui, China
- Department of Dermatology, Shannan People's Hospital, Shannan, China
- First Clinical Medical College, Anhui Medical University, Hefei, Anhui, China
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of Pharmacology, Basic Medical College, Anhui Medical University, Hefei, Anhui, China
| | - Qiuyu Liu
- First Clinical Medical College, Anhui Medical University, Hefei, Anhui, China
| | - Naiyu Ke
- Department of Ophthalmology, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Hao Li
- Department of Urology, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Feiyang Sun
- First Clinical Medical College, Anhui Medical University, Hefei, Anhui, China
| | - Jiali Zhang
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, Anhui, China
- Department of Skin Genetics, Anhui Province Laboratory of Inflammation and Immune Mediated Diseases, Hefei, Anhui, China
- Department of Dermatology, Shannan People's Hospital, Shannan, China
| | - Zhengwei Zhu
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, Anhui, China
- Department of Skin Genetics, Anhui Province Laboratory of Inflammation and Immune Mediated Diseases, Hefei, Anhui, China
- Department of Dermatology, Shannan People's Hospital, Shannan, China
| |
Collapse
|
43
|
Alves VC, Carro E, Figueiro-Silva J. Unveiling DNA methylation in Alzheimer's disease: a review of array-based human brain studies. Neural Regen Res 2024; 19:2365-2376. [PMID: 38526273 PMCID: PMC11090417 DOI: 10.4103/1673-5374.393106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 12/05/2023] [Indexed: 03/26/2024] Open
Abstract
The intricacies of Alzheimer's disease pathogenesis are being increasingly illuminated by the exploration of epigenetic mechanisms, particularly DNA methylation. This review comprehensively surveys recent human-centered studies that investigate whole genome DNA methylation in Alzheimer's disease neuropathology. The examination of various brain regions reveals distinctive DNA methylation patterns that associate with the Braak stage and Alzheimer's disease progression. The entorhinal cortex emerges as a focal point due to its early histological alterations and subsequent impact on downstream regions like the hippocampus. Notably, ANK1 hypermethylation, a protein implicated in neurofibrillary tangle formation, was recurrently identified in the entorhinal cortex. Further, the middle temporal gyrus and prefrontal cortex were shown to exhibit significant hypermethylation of genes like HOXA3, RHBDF2, and MCF2L, potentially influencing neuroinflammatory processes. The complex role of BIN1 in late-onset Alzheimer's disease is underscored by its association with altered methylation patterns. Despite the disparities across studies, these findings highlight the intricate interplay between epigenetic modifications and Alzheimer's disease pathology. Future research efforts should address methodological variations, incorporate diverse cohorts, and consider environmental factors to unravel the nuanced epigenetic landscape underlying Alzheimer's disease progression.
Collapse
Affiliation(s)
- Victoria Cunha Alves
- Neurodegenerative Diseases Group, Hospital Universitario 12 de Octubre Research Institute (imas12), Madrid, Spain
- Network Center for Biomedical Research, Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- PhD Program in Neuroscience, Autonoma de Madrid University, Madrid, Spain
- Neurotraumatology and Subarachnoid Hemorrhage Group, Hospital Universitario 12 de Octubre Research Institute (imas12), Madrid, Spain
| | - Eva Carro
- Network Center for Biomedical Research, Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Neurobiology of Alzheimer's Disease Unit, Functional Unit for Research Into Chronic Diseases, Instituto de Salud Carlos III, Madrid, Spain
| | - Joana Figueiro-Silva
- Institute of Medical Genetics, University of Zurich, Zurich, Switzerland
- Department of Molecular Life Science, University of Zurich, Zurich, Switzerland
| |
Collapse
|
44
|
Li J, Yushanjiang F, Fang Z, Liu W. NAT10-mediated RNA ac4C acetylation contributes to the myocardial infarction-induced cardiac fibrosis. J Cell Mol Med 2024; 28:e70141. [PMID: 39482983 PMCID: PMC11528131 DOI: 10.1111/jcmm.70141] [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: 09/22/2023] [Revised: 08/31/2024] [Accepted: 10/01/2024] [Indexed: 11/03/2024] Open
Abstract
Cardiac fibrosis is featured cardiac fibroblast activation and extracellular matrix accumulation. Ac4C acetylation is an important epigenetic regulation of RNAs that has been recently discovered, and it is solely carried out by NAT10, the exclusive enzyme used for the modification. However, the potential regulatory mechanisms of ac4C acetylation in myocardial fibrosis following myocardial infarction remain poorly understood. In our study, we activated fibroblasts in vitro using TGF-β1 (20 ng/mL), followed by establishing a myocardial infarction mouse model to evaluate the impact of NAT10 on collagen synthesis and cardiac fibroblast proliferation. We utilized a NAT10 inhibitor, Remodelin, to attenuate the acetylation capacity of NAT10. In the cardiac fibrosis tissues of chronic myocardial infarction mice and cultured cardiac fibroblasts (CFs) in response to TGF-β1 treatment, there was an elevation in the levels of NAT10 expression. This increase facilitated proliferation, the accumulation of collagens, as well as fibroblast-to-myofibroblast transition. Through the administration of Remodelin, we effectively reduced cardiac fibrosis in myocardial infarction mice by inhibiting NAT10's ability to acetylate mRNA. Inhibition of NAT10 resulted in changes in collagen-related gene expression and ac4C acetylation levels. Mechanistically, we found that NAT10 upregulates the acetylation modification of BCL-XL mRNA and enhances the stability of BCL-XL mRNA, thereby upregulating its protein expression, inhibiting the activation of Caspase3 and blocking the apoptosis of CFs. Therefore, the crucial involvement of NAT10-mediated ac4C acetylation is significant in the cardiac fibrosis progression, affording promising molecular targets for the treatment of fibrosis and relevant cardiac diseases.
Collapse
Affiliation(s)
- Jun Li
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanHubeiChina
- Cardiovascular Research InstituteWuhan UniversityWuhanHubeiChina
- Hubei Key Laboratory of CardiologyWuhanHubeiChina
| | - Feierkaiti Yushanjiang
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanHubeiChina
- Cardiovascular Research InstituteWuhan UniversityWuhanHubeiChina
- Hubei Key Laboratory of CardiologyWuhanHubeiChina
| | - Zhao Fang
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanHubeiChina
- Cardiovascular Research InstituteWuhan UniversityWuhanHubeiChina
- Hubei Key Laboratory of CardiologyWuhanHubeiChina
| | - Wan‐li Liu
- Department of Pediatric, Maternal and Child Health Hospital of Hubei Province, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiChina
| |
Collapse
|
45
|
Qi Y, Li T, Zhou Y, Hao Y, Zhang J. RNA modification regulators as promising biomarkers in gynecological cancers. Cell Biol Toxicol 2024; 40:92. [PMID: 39472384 PMCID: PMC11522084 DOI: 10.1007/s10565-024-09924-y] [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: 04/10/2024] [Accepted: 10/02/2024] [Indexed: 11/02/2024]
Abstract
This review explores the evolving landscape of gynecological oncology by focusing on emerging RNA modification signatures as promising biomarkers for assessing the risk and progression of ovarian, cervical, and uterine cancers. It provides a comprehensive overview of common RNA modifications, especially m6A, and their roles in cellular processes, emphasizing their implications in gynecological cancer development. The review meticulously examines specific m6A regulators including "writers", "readers", and "erasers" associated with three gynecological cancer types, discussing their involvement in initiation and progression. Methodologies for detecting RNA modifications are surveyed, highlighting advancements in high-throughput techniques with high sensitivity. A critical analysis of studies identifying m6A regulators as potential biomarkers is presented, addressing their diagnostic or prognostic significance. Mechanistic insights into RNA modification-mediated cancer progression are explored, shedding light on molecular pathways and potential therapeutic targets. Despite current challenges, the review discusses ongoing research efforts, future directions, and the transformative possibility of RNA modifications on early assessment and personalized therapy in gynecological oncology.
Collapse
Affiliation(s)
- Yue Qi
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, 36 Sanhao St, Shenyang, 110004, Liaoning, China
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, Xi'an, Shanxi, China.
| | - Yang Zhou
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, 36 Sanhao St, Shenyang, 110004, Liaoning, China
| | - Yingying Hao
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, 36 Sanhao St, Shenyang, 110004, Liaoning, China.
| | - Jin Zhang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, 36 Sanhao St, Shenyang, 110004, Liaoning, China.
| |
Collapse
|
46
|
Xu Z, Zheng X, Fan J, Jiao Y, Huang S, Xie Y, Xu S, Lu Y, Liu A, Liu R, Yang Y, Luo GZ, Pan T, Wang X. Microbiome-induced reprogramming in post-transcriptional landscape using nanopore direct RNA sequencing. Cell Rep 2024; 43:114798. [PMID: 39365698 DOI: 10.1016/j.celrep.2024.114798] [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: 05/07/2024] [Revised: 08/10/2024] [Accepted: 09/10/2024] [Indexed: 10/06/2024] Open
Abstract
It has been widely recognized that the microbiota has the capacity to shape host gene expression and physiological functions. However, there remains a paucity of comprehensive study revealing the host transcriptional landscape regulated by the microbiota. Here, we comprehensively examined mRNA landscapes in mouse tissues (brain and cecum) from specific-pathogen-free and germ-free mice using nanopore direct RNA sequencing. Our results show that the microbiome has global influence on a host's RNA modifications (m6A, m5C, Ψ), isoform generation, poly(A) tail length, and transcript abundance in both brain and cecum tissues. Moreover, the microbiome exerts tissue-specific effects on various post-transcriptional regulatory processes. In addition, the microbiome impacts the coordination of multiple RNA modifications in host brain and cecum tissues. In conclusion, we establish the relationship between microbial regulation and gene expression. Our results help the understanding of the mechanisms by which the microbiome reprograms host gene expression.
Collapse
Affiliation(s)
- Zihe Xu
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Xiaoqi Zheng
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Jiajun Fan
- School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Yuting Jiao
- School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Sihao Huang
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL 60637, USA
| | - Yingyuan Xie
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
| | - Shunlan Xu
- School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Yi Lu
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Anrui Liu
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Runzhou Liu
- School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Ying Yang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
| | - Guan-Zheng Luo
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
| | - Tao Pan
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL 60637, USA
| | - Xiaoyun Wang
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; School of Life Sciences, South China Normal University, Guangzhou 510631, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
47
|
Yao J, Yao P, Li Y, He K, Ma X, Yang Q, Jia J, Chen Z, Yu S, Gu S, Chen K, Zhao Y, Li W, Wang G, Guo M. Integration of multi-omics data revealed the orphan CpG islands and enhancer-dominated c is-regulatory network in glioma. iScience 2024; 27:110946. [PMID: 39391717 PMCID: PMC11465130 DOI: 10.1016/j.isci.2024.110946] [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: 03/18/2024] [Revised: 07/12/2024] [Accepted: 09/10/2024] [Indexed: 10/12/2024] Open
Abstract
The complex transcriptional regulatory network leads to the poor prognosis of glioma. The role of orphan CpG islands (oCGIs) in the transcriptional regulatory network has been overlooked. We conducted a comprehensive exploration of the cis-regulatory roles of oCGIs and enhancers by integrating multi-omics data. Direct regulation of target genes by oCGIs or enhancers is of great importance in the cis-regulatory network. Furthermore, based on single-cell multi-omics data, we found that the highly activated cis-regulatory network in cluster 2 (C2) sustains the high proliferative potential of glioma cells. The upregulation of oCGIs and enhancers related genes in C2 results in glioma patients exhibiting resistance to radiotherapy and chemotherapy. These findings were further validated through glioma cell line related experiments. Our study offers insight into the pathogenesis of glioma and provides a strategy to treat this challenging disease.
Collapse
Affiliation(s)
- Jiawei Yao
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Penglei Yao
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Yang Li
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Ke He
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Xinqi Ma
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Qingsong Yang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Junming Jia
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Zeren Chen
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Shan Yu
- Department of Pathology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Shuqing Gu
- Department of Neurosurgery, The First Hospital of Qiqihar, Qiqihar 161005, China
| | - Kunliang Chen
- Department of Neurosurgery, People’s Hospital of the Daxing’an Mountain Range, Daxing’an Mountain Range 165300, China
| | - Yan Zhao
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Weihua Li
- Medical Imaging Department, Shenzhen Second People’s Hospital, the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen 518035, China
| | - Guangzhi Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Mian Guo
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| |
Collapse
|
48
|
Ren X, Feng Z, Ma X, Huo L, Zhou H, Bai A, Feng S, Zhou Y, Weng X, Fan C. m6A/m1A/m5C-Associated Methylation Alterations and Immune Profile in MDD. Mol Neurobiol 2024; 61:8000-8025. [PMID: 38453794 DOI: 10.1007/s12035-024-04042-6] [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: 10/17/2023] [Accepted: 02/07/2024] [Indexed: 03/09/2024]
Abstract
Major depressive disorder (MDD) is a prevalent psychiatric condition often accompanied by severe impairments in cognitive and functional capacities. This research was conducted to identify RNA modification-related gene signatures and associated functional pathways in MDD. Differentially expressed RNA modification-related genes in MDD were first identified. And a random forest model was developed and distinct RNA modification patterns were discerned based on signature genes. Then, comprehensive analyses of RNA modification-associated genes in MDD were performed, including functional analyses and immune cell infiltration. The study identified 29 differentially expressed RNA modification-related genes in MDD and two distinct RNA modification patterns. TRMT112, MBD3, NUDT21, and IGF2BP1 of the risk signature were detected. Functional analyses confirmed the involvement of RNA modification in pathways like phosphatidylinositol 3-kinase signaling and nucleotide oligomerization domain (NOD)-like receptor signaling in MDD. NUDT21 displayed a strong positive correlation with type 2 T helper cells, while IGF2BP1 negatively correlated with activated CD8 T cells, central memory CD4 T cells, and natural killer T cells. In summary, further research into the roles of NUDT21 and IGF2BP1 would be valuable for understanding MDD prognosis. The identified RNA modification-related gene signatures and pathways provide insights into MDD molecular etiology and potential diagnostic biomarkers.
Collapse
Affiliation(s)
- Xin Ren
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, 55 Zhongshan Avenue West, Tianhe District, Guangzhou, 510631, China
- Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, 510631, China
| | - Zhuxiao Feng
- Department of Psychiatry, Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, China
| | - Xiaodong Ma
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, 55 Zhongshan Avenue West, Tianhe District, Guangzhou, 510631, China
- Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, 510631, China
| | - Lijuan Huo
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, 55 Zhongshan Avenue West, Tianhe District, Guangzhou, 510631, China
- Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, 510631, China
| | - Huiying Zhou
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, 55 Zhongshan Avenue West, Tianhe District, Guangzhou, 510631, China
- Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, 510631, China
| | - Ayu Bai
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, 55 Zhongshan Avenue West, Tianhe District, Guangzhou, 510631, China
- Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, 510631, China
| | - Shujie Feng
- Department of Rehabilitation Medicine, Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, China
| | - Ying Zhou
- Department of Psychiatry, Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, China
| | - Xuchu Weng
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, 55 Zhongshan Avenue West, Tianhe District, Guangzhou, 510631, China.
- Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, 510631, China.
| | - Changhe Fan
- Department of Psychiatry, Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, China.
| |
Collapse
|
49
|
Park HH, Armstrong MJ, Gorin FA, Lein PJ. Air Pollution as an Environmental Risk Factor for Alzheimer's Disease and Related Dementias. MEDICAL RESEARCH ARCHIVES 2024; 12:5825. [PMID: 39822906 PMCID: PMC11736697 DOI: 10.18103/mra.v12i10.5825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Alzheimer's disease and related dementias are a leading cause of morbidity in our aging populations. Although influenced by genetic factors, fewer than 5% of Alzheimer's disease and related dementia cases are due solely to genetic causes. There is growing scientific consensus that these dementias arise from complex gene by environment interactions. The 2020 Lancet Commission on dementia prevention, intervention, and care identified 12 modifiable risk factors of dementia, including lifestyle, educational background, comorbidities, and environmental exposures to environmental contaminants. In this review, we summarize the current understanding and data gaps regarding the role(s) of environmental pollutants in the etiology of Alzheimer's disease and related dementias with a focus on air pollution. In addition to summarizing findings from epidemiological and experimental animal studies that link airborne exposures to environmental contaminants to increased risk and/or severity of Alzheimer's disease and related dementias, we discuss currently hypothesized mechanism(s) underlying these associations, including peripheral inflammation, neuroinflammation and epigenetic changes. Key data gaps in this rapidly expanding investigative field and approaches for addressing these gaps are also addressed.
Collapse
Affiliation(s)
- Heui Hye Park
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
| | - Matthew J. Armstrong
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
| | - Fredric A. Gorin
- Department of Molecular Biosciences, School of Veterinary Medicine, and Department of Neurology, School of Medicine, University of California, Davis, CA 95616, USA
| | - Pamela J. Lein
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
| |
Collapse
|
50
|
Cheng L, Wang C, Zhao D, Wu S. Narrative review of research progress of RNA m 5C methylation in head and neck malignancies. Transl Cancer Res 2024; 13:5112-5122. [PMID: 39430837 PMCID: PMC11483327 DOI: 10.21037/tcr-24-103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 08/11/2024] [Indexed: 10/22/2024]
Abstract
Background and Objective Head and neck malignancies encompass a spectrum of malignant tumors occurring in the head and neck region, characterized by rapid progression, high recurrence rates, and dismal prognoses. Despite significant advancements in comprehensive surgery-based therapies, the 5-year survival rate for patients has not shown substantial improvement. There is an urgent need to investigate novel targeted therapies. With the advancements in epigenetics, RNA 5-methylcytosine (m5C) methylation, a prevalent form of RNA modification, has been identified by numerous studies as playing a pivotal role in the pathological processes of tumorigenesis and development. However, a comprehensive review within the realm of head and neck malignancies is currently lacking. This study aims to comprehensively review the biological implications of RNA m5C methylation regulators in the pathogenesis and progression of various systemic malignant tumors, with a specific focus on exploring the potential impact of RNA m5C methylation on head and neck malignancies. Methods A literature search on RNA m5C methylation and head and neck malignancies was conducted using PubMed, resulting in the inclusion of 46 relevant articles. The Cancer Genome Atlas (TCGA) database was utilized to analyze the correlation between m5C regulatory factors and clinicopathological features in patients with head and neck squamous cell carcinoma (HNSCC). Key Content and Findings Aberrant expression of RNA m5C methylation regulators is observed in head and neck malignancies, displaying a correlation with the clinicopathological grading of tumors. Conclusions RNA m5C methylation may contribute to the progression of head and neck malignancies and could be associated with an unfavorable prognosis for patients. These findings offer valuable insights for the development of targeted treatments for head and neck malignancies.
Collapse
Affiliation(s)
- Lang Cheng
- The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, China
- School of Stomatology, Southwest Medical University, Luzhou, China
| | - Chengtao Wang
- The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, China
- School of Stomatology, Southwest Medical University, Luzhou, China
| | - Dan Zhao
- The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, China
- School of Stomatology, Southwest Medical University, Luzhou, China
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Luzhou, China
| | - Shuangjiang Wu
- The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, China
- School of Stomatology, Southwest Medical University, Luzhou, China
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Luzhou, China
| |
Collapse
|