1
|
Lan Y, Xia Z, Shao Q, Lin P, Lu J, Xiao X, Zheng M, Chen D, Dou Y, Xie Q. Synonymous mutations promote tumorigenesis by disrupting m 6A-dependent mRNA metabolism. Cell 2025; 188:1828-1841.e15. [PMID: 39952247 DOI: 10.1016/j.cell.2025.01.026] [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: 06/03/2024] [Revised: 11/04/2024] [Accepted: 01/17/2025] [Indexed: 02/17/2025]
Abstract
Cancer cells acquire numerous mutations during tumorigenesis, including synonymous mutations that do not change the amino acid sequence of a protein. RNA N6-methyladenosine (m6A) is a post-transcriptional modification that plays critical roles in oncogenesis. Herein, we identified 12,849 mutations in the cancer genome with the potential to perturb m6A modification patterns, which we refer to as "m6A disruption mutations (m6A-DMs)." These are either synonymous m6A-DMs (sm6A-DMs) or missense m6A-DMs (mm6A-DMs) mutations, and the former is enriched within tumor suppressor genes, such as CDKN2A and BRCA2. Using epitranscriptomic editing, we demonstrate that manipulating m6A levels at specific sm6A-DM sites influences mRNA stability. Furthermore, introducing CDKN2A sm6A-DMs into cancer cells promotes tumor growth while BRCA2 sm6A-DMs sensitize tumors to the poly (ADP-ribose) polymerase inhibitor (PARPi) treatment. Our findings demonstrate sm6A-DMs as potential oncogenic drivers, unveiling implications for synonymous mutations in tumorigenesis and beyond.
Collapse
Affiliation(s)
- Yiheng Lan
- Westlake Disease Modeling Laboratory, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310024, China; School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China
| | - Zhen Xia
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China
| | - Qizhe Shao
- Center for Regeneration and Cell Therapy of Zhejiang University, University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Peng Lin
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China
| | - Jinhong Lu
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China; Fudan University, Shanghai 200433, China
| | - Xiaoying Xiao
- Center for Regeneration and Cell Therapy of Zhejiang University, University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Mengyue Zheng
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China
| | - Di Chen
- Center for Regeneration and Cell Therapy of Zhejiang University, University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China.
| | - Yanmei Dou
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310024, China.
| | - Qi Xie
- Westlake Disease Modeling Laboratory, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310024, China; School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China.
| |
Collapse
|
2
|
Zou WW, Liao YF, Huang XR, Lin WS, Lu SM. Circular RNA circCD44 targets methyltransferase-like 3/homeobox containing 1 to facilitate esophageal squamous cell carcinoma progression. Kaohsiung J Med Sci 2025; 41:e12950. [PMID: 40008841 DOI: 10.1002/kjm2.12950] [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: 11/14/2024] [Revised: 01/07/2025] [Accepted: 01/22/2025] [Indexed: 02/27/2025] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is a prevalent gastrointestinal malignancy with unclear functional mechanisms of circular RNAs. This study focused on investigating the role of circCD44 in ESCC progression. The expression of circCD44 and homeobox containing 1 (HMBOX1) was found to be elevated in ESCC cell lines. The overexpression of circCD44 or HMBOX1 enhanced proliferation and invasion, while inhibiting apoptosis in ESCC cells, whereas knockdown of these genes had inverse effects. METTL3 was observed to bind to circCD44 and HMBOX1 mRNA, promoting m6A modification in HMBOX1 mRNA and subsequent HMBOX1 protein expression. Knockdown of METTL3 or HMBOX1 attenuated the oncogenic effects of circCD44 overexpression both in vitro and in vivo. These findings suggest that circCD44 promotes ESCC progression by facilitating HMBOX1 mRNA m6A modification and protein expression through METTL3 binding, providing insights into the molecular mechanisms underlying ESCC pathogenesis.
Collapse
Affiliation(s)
- Wei-Wen Zou
- Department of Pathology, Longgang Center Hospital of Shenzhen, Shenzhen, Guangdong, PR China
| | - Yan-Fang Liao
- Department of Pathology, Longgang Center Hospital of Shenzhen, Shenzhen, Guangdong, PR China
| | - Xi-Rui Huang
- Department of Pathology, Longgang Center Hospital of Shenzhen, Shenzhen, Guangdong, PR China
| | - Wen-Song Lin
- Department of Pathology, Longgang Center Hospital of Shenzhen, Shenzhen, Guangdong, PR China
| | - Shan-Ming Lu
- Department of Pathology, Longgang Center Hospital of Shenzhen, Shenzhen, Guangdong, PR China
| |
Collapse
|
3
|
Pilala KM, Panoutsopoulou K, Papadimitriou MA, Soureas K, Scorilas A, Avgeris M. Exploring the methyl-verse: Dynamic interplay of epigenome and m6A epitranscriptome. Mol Ther 2025; 33:447-464. [PMID: 39659016 PMCID: PMC11852398 DOI: 10.1016/j.ymthe.2024.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 11/19/2024] [Accepted: 12/05/2024] [Indexed: 12/12/2024] Open
Abstract
The orchestration of dynamic epigenetic and epitranscriptomic modifications is pivotal for the fine-tuning of gene expression. However, these modifications are traditionally examined independently. Recent compelling studies have disclosed an interesting communication and interplay between m6A RNA methylation (m6A epitranscriptome) and epigenetic modifications, enabling the formation of feedback circuits and cooperative networks. Intriguingly, the interaction between m6A and DNA methylation machinery, coupled with the crosstalk between m6A RNA and histone modifications shape the transcriptional profile and translational efficiency. Moreover, m6A modifications interact also with non-coding RNAs, modulating their stability, abundance, and regulatory functions. In the light of these findings, m6A imprinting acts as a versatile checkpoint, linking epigenetic and epitranscriptomic layers toward a multilayer and time-dependent control of gene expression and cellular homeostasis. The scope of the present review is to decipher the m6A-coordinated circuits with DNA imprinting, chromatin architecture, and non-coding RNAs networks in normal physiology and carcinogenesis. Ultimately, we summarize the development of innovative CRISPR-dCas engineering platforms fused with m6A catalytic components (m6A writers or erasers) to achieve transcript-specific editing of m6A epitranscriptomes that can create new insights in modern RNA therapeutics.
Collapse
Affiliation(s)
- Katerina-Marina Pilala
- Department of Biochemistry and Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Konstantina Panoutsopoulou
- Department of Biochemistry and Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria-Alexandra Papadimitriou
- Department of Biochemistry and Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Konstantinos Soureas
- Department of Biochemistry and Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece; Laboratory of Clinical Biochemistry - Molecular Diagnostics, Second Department of Pediatrics, School of Medicine, National and Kapodistrian University of Athens, "P. & A. Kyriakou" Children's Hospital, Athens, Greece
| | - Andreas Scorilas
- Department of Biochemistry and Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Margaritis Avgeris
- Department of Biochemistry and Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece; Laboratory of Clinical Biochemistry - Molecular Diagnostics, Second Department of Pediatrics, School of Medicine, National and Kapodistrian University of Athens, "P. & A. Kyriakou" Children's Hospital, Athens, Greece.
| |
Collapse
|
4
|
Xie X, Fang Z, Zhang H, Wang Z, Li J, Jia Y, Shang L, Cao F, Li F. The role of N(6)-methyladenosine (m6a) modification in cancer: recent advances and future directions. EXCLI JOURNAL 2025; 24:113-150. [PMID: 39967906 PMCID: PMC11830918 DOI: 10.17179/excli2024-7935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 12/18/2024] [Indexed: 02/20/2025]
Abstract
N(6)-methyladenosine (m6A) modification is the most abundant and prevalent internal modification in eukaryotic mRNAs. The role of m6A modification in cancer has become a hot research topic in recent years and has been widely explored. m6A modifications have been shown to regulate cancer occurrence and progression by modulating different target molecules. This paper reviews the recent research progress of m6A modifications in cancer and provides an outlook on future research directions, especially the development of molecularly targeted drugs. See also the graphical abstract(Fig. 1).
Collapse
Affiliation(s)
- Xiaozhou Xie
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Zhen Fang
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Haoyu Zhang
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Zheng Wang
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jie Li
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yuchen Jia
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Liang Shang
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Feng Cao
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Fei Li
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
5
|
Cui Z, Liu X, E T, Lin H, Wang D, Liu Y, Ruan X, Wang P, Liu L, Xue Y. Effect of SNORD113-3/ADAR2 on glycolipid metabolism in glioblastoma via A-to-I editing of PHKA2. Cell Mol Biol Lett 2025; 30:5. [PMID: 39794701 PMCID: PMC11724473 DOI: 10.1186/s11658-024-00680-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 12/17/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is a highly aggressive brain tumor, characterized by its poor prognosis. Glycolipid metabolism is strongly associated with GBM development and malignant behavior. However, the precise functions of snoRNAs and ADARs in glycolipid metabolism within GBM cells remain elusive. The objective of the present study is to delve into the underlying mechanisms through which snoRNAs and ADARs exert regulatory effects on glycolipid metabolism in GBM cells. METHODS RNA immunoprecipitation and RNA pull-down experiments were conducted to verify the homodimerization of ADAR2 by SNORD113-3, and Sanger sequencing and Western blot experiments were used to detect the A-to-I RNA editing of PHKA2 mRNA by ADAR2. Furthermore, the phosphorylation of EBF1 was measured by in vitro kinase assay. Finally, in vivo studies using nude mice confirmed that SNORD113-3 and ADAR2 overexpression, along with PHKA2 knockdown, could suppress the formation of subcutaneous xenograft tumors and improve the outcome of tumor-bearing nude mice. RESULTS We found that PHKA2 in GBM significantly promoted glycolipid metabolism, while SNORD113-3, ADAR2, and EBF1 significantly inhibited glycolipid metabolism. SNORD113-3 promotes ADAR2 protein expression by promoting ADAR2 homodimer formation. ADAR2 mediates the A-to-I RNA editing of PHKA2 mRNA. Mass spectrometry analysis and in vitro kinase testing revealed that PHKA2 phosphorylates EBF1 on Y256, reducing the stability and expression of EBF1. Furthermore, direct binding of EBF1 to PKM2 and ACLY promoters was observed, suggesting the inhibition of their expression by EBF1. These findings suggest the existence of a SNORD113-3/ADAR2/PHKA2/EBF1 pathway that collectively regulates the metabolism of glycolipid and the growth of GBM cells. Finally, in vivo studies using nude mice confirmed that knockdown of PHKA2, along with overexpression of SNORD113-3 and ADAR2, could obviously suppress GBM subcutaneous xenograft tumor formation and improve the outcome of those tumor-bearing nude mice. CONCLUSIONS Herein, we clarified the underlying mechanism involving the SNORD113-3/ADAR2/PHKA2/EBF1 pathway in the regulation of GBM cell growth and glycolipid metabolism. Our results provide a framework for the development of innovative therapeutic interventions to improve the prognosis of patients with GBM.
Collapse
Affiliation(s)
- Zheng Cui
- Department of Neurology, The First Affiliated Hospital, China Medical University, Shenyang, 110001, Liaoning, China
- Key Laboratory of Neurological Disease Big Data of Liaoning Province, Shenyang, China
- Shenyang Clinical Medical Research Center for Difficult and Serious Diseases of the Nervous System, Shenyang, China
| | - Xiaobai Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, China
- Key Laboratory of Neuro-Oncology in Liaoning Province, Shenyang, 110004, China
- Liaoning Medical Surgery and Rehabilitation Robot Technology Engineering Research Center, Shenyang, 110004, China
| | - Tiange E
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, China
- Key Laboratory of Neuro-Oncology in Liaoning Province, Shenyang, 110004, China
- Liaoning Medical Surgery and Rehabilitation Robot Technology Engineering Research Center, Shenyang, 110004, China
| | - Hongda Lin
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, China
- Key Laboratory of Neuro-Oncology in Liaoning Province, Shenyang, 110004, China
- Liaoning Medical Surgery and Rehabilitation Robot Technology Engineering Research Center, Shenyang, 110004, China
| | - Di Wang
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, China
- Key Laboratory of Neuro-Oncology in Liaoning Province, Shenyang, 110004, China
- Liaoning Medical Surgery and Rehabilitation Robot Technology Engineering Research Center, Shenyang, 110004, China
| | - Yunhui Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, China
- Key Laboratory of Neuro-Oncology in Liaoning Province, Shenyang, 110004, China
- Liaoning Medical Surgery and Rehabilitation Robot Technology Engineering Research Center, Shenyang, 110004, China
| | - Xuelei Ruan
- Key Laboratory of Neuro-Oncology in Liaoning Province, Shenyang, 110004, China
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, China
| | - Ping Wang
- Key Laboratory of Neuro-Oncology in Liaoning Province, Shenyang, 110004, China
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, China
| | - Libo Liu
- Key Laboratory of Neuro-Oncology in Liaoning Province, Shenyang, 110004, China
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, China
| | - Yixue Xue
- Key Laboratory of Neuro-Oncology in Liaoning Province, Shenyang, 110004, China.
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, China.
| |
Collapse
|
6
|
Liu G, Wang Y, He Y, Yu M, Liu X, Wang F. Harnessing Demethylase-Regulated Catalytic DNA Circuit for In-Situ Investigation of the Regulatory Connection with MicroRNA. Anal Chem 2024; 96:20304-20311. [PMID: 39668155 DOI: 10.1021/acs.analchem.4c05225] [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: 12/14/2024]
Abstract
Insight into the epigenetic modulation-correlated molecule interactions has significant implications for the in-depth understanding of intracellular intricate biological networks. However, there is currently a lack of reliable biological tools for elucidating the potential correlation between epigenetic regulators and relevant genes, e.g., microRNAs (miRNAs). Herein, an alkB homologue 5 (ALKBH5, a key epigenetic regulator)-modulated catalytic DNA circuit (ACD) was constructed by grafting a N6-methyladenosine (m6A)-caged I-R3 DNAzyme into the circuitry components for achieving the on-site miRNA imaging in living cells. Specifically, the catalytic activity of I-R3 DNAzyme could be effectively suppressed by the m6A modification situated at its highly sequence-conserved core region and then be selectively restored through the ALKBH5-mediated demethylation pathway. And the ALKBH5-activated I-R3 DNAzyme allowed the highly efficient DNA cleaving reaction in the presence of DNAzyme cofactors, resulting in the liberation of catalytic hairpin assembly (CHA) reactants. Subsequently, target miRNA triggered the CHA circuit to produce a duplex DNA product while releasing the miRNA analyte. The liberated miRNA could autonomously trigger the next round of the CHA assembly cycle for generating the amplified fluorescence readout. By virtue of the stimuli-responsive activation and the CHA amplification circuit, the ACD system achieved highly specific and sensitive imaging of miRNA in tumor cells. Moreover, this efficiently and reliably ALKBH5-activated DNA circuit is demonstrated to reveal the underlying relationship between activator ALKBH5 and miRNA. Overall, the developed ACD system provides a promising tool for the robust on-site profiling of epigenetic-involved signal pathways, thus displaying great potential in bioanalytical applications.
Collapse
Affiliation(s)
- Guangqin Liu
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, People's Republic of China
- Department of Gastroenterology, Hubei Key Laboratory of Tumor Biological Behavior, Zhongnan Hospital of Wuhan University, Wuhan 430072, People's Republic of China
| | - Yifei Wang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, People's Republic of China
- Department of Gastroenterology, Hubei Key Laboratory of Tumor Biological Behavior, Zhongnan Hospital of Wuhan University, Wuhan 430072, People's Republic of China
| | - Yuqiu He
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, People's Republic of China
- Department of Gastroenterology, Hubei Key Laboratory of Tumor Biological Behavior, Zhongnan Hospital of Wuhan University, Wuhan 430072, People's Republic of China
| | - Mengdi Yu
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, People's Republic of China
- Department of Gastroenterology, Hubei Key Laboratory of Tumor Biological Behavior, Zhongnan Hospital of Wuhan University, Wuhan 430072, People's Republic of China
| | - Xiaoqing Liu
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, People's Republic of China
- Department of Gastroenterology, Hubei Key Laboratory of Tumor Biological Behavior, Zhongnan Hospital of Wuhan University, Wuhan 430072, People's Republic of China
| | - Fuan Wang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, People's Republic of China
- Department of Gastroenterology, Hubei Key Laboratory of Tumor Biological Behavior, Zhongnan Hospital of Wuhan University, Wuhan 430072, People's Republic of China
- Research Institute of Shenzhen, Wuhan University, Shenzhen 518057, People's Republic of China
| |
Collapse
|
7
|
Kaur P, Sharma P, Bhatia P, Singh M. Current insights on m6A RNA modification in acute leukemia: therapeutic targets and future prospects. Front Oncol 2024; 14:1445794. [PMID: 39600630 PMCID: PMC11590065 DOI: 10.3389/fonc.2024.1445794] [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: 06/08/2024] [Accepted: 10/08/2024] [Indexed: 11/29/2024] Open
Abstract
RNA modification is the critical mechanism for regulating post-transcriptional processes. There are more than 150 RNA modifications reported so far, among which N6-Methyladenosine is the most prevalent one. M6A RNA modification complex consists of 'writers', 'readers' and 'erasers' which together in a group catalyze, recognize and regulate the methylation process of RNA and thereby regulate the stability and translation of mRNA. The discovery of erasers also known as demethylases, revolutionized the research on RNA modifications as it revealed that this modification is reversible. Since then, various studies have focused on discovering the role of m6A modification in various diseases especially cancers. Aberrant expression of these 'readers', 'writers', and 'erasers' is found to be altered in various cancers resulting in disturbance of cellular homeostasis. Acute leukemias are the most common cancer found in pediatric patients and account for 20% of adult cases. Dysregulation of the RNA modifying complex have been reported in development and progression of hematopoietic malignancies. Further, targeting m6A modification is the new approach for cancer immunotherapy and is being explored extensively. This review provides detailed information about current information on the role of m6A RNA modification in acute leukemia and their therapeutic potential.
Collapse
Affiliation(s)
| | | | | | - Minu Singh
- Haematology-Oncology Unit, Department of Paediatrics, Postgraduate Institute of Medical
Education and Research, Chandigarh, India
| |
Collapse
|
8
|
Nabeel-Shah S, Pu S, Burke GL, Ahmed N, Braunschweig U, Farhangmehr S, Lee H, Wu M, Ni Z, Tang H, Zhong G, Marcon E, Zhang Z, Blencowe BJ, Greenblatt JF. Recruitment of the m 6A/m6Am demethylase FTO to target RNAs by the telomeric zinc finger protein ZBTB48. Genome Biol 2024; 25:246. [PMID: 39300486 PMCID: PMC11414060 DOI: 10.1186/s13059-024-03392-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 09/11/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND N6-methyladenosine (m6A), the most abundant internal modification on eukaryotic mRNA, and N6, 2'-O-dimethyladenosine (m6Am), are epitranscriptomic marks that function in multiple aspects of posttranscriptional regulation. Fat mass and obesity-associated protein (FTO) can remove both m6A and m6Am; however, little is known about how FTO achieves its substrate selectivity. RESULTS Here, we demonstrate that ZBTB48, a C2H2-zinc finger protein that functions in telomere maintenance, associates with FTO and binds both mRNA and the telomere-associated regulatory RNA TERRA to regulate the functional interactions of FTO with target transcripts. Specifically, depletion of ZBTB48 affects targeting of FTO to sites of m6A/m6Am modification, changes cellular m6A/m6Am levels and, consequently, alters decay rates of target RNAs. ZBTB48 ablation also accelerates growth of HCT-116 colorectal cancer cells and modulates FTO-dependent regulation of Metastasis-associated protein 1 (MTA1) transcripts by controlling the binding to MTA1 mRNA of the m6A reader IGF2BP2. CONCLUSIONS Our findings thus uncover a previously unknown mechanism of posttranscriptional regulation in which ZBTB48 co-ordinates RNA-binding of the m6A/m6Am demethylase FTO to control expression of its target RNAs.
Collapse
Affiliation(s)
- Syed Nabeel-Shah
- Donnelly Centre, University of Toronto, Toronto, M5S 3E1, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, M5S 1A8, Canada
| | - Shuye Pu
- Donnelly Centre, University of Toronto, Toronto, M5S 3E1, Canada
| | - Giovanni L Burke
- Donnelly Centre, University of Toronto, Toronto, M5S 3E1, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, M5S 1A8, Canada
| | - Nujhat Ahmed
- Donnelly Centre, University of Toronto, Toronto, M5S 3E1, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, M5S 1A8, Canada
| | | | - Shaghayegh Farhangmehr
- Donnelly Centre, University of Toronto, Toronto, M5S 3E1, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, M5S 1A8, Canada
| | - Hyunmin Lee
- Donnelly Centre, University of Toronto, Toronto, M5S 3E1, Canada
- Department of Computer Sciences, University of Toronto, Toronto, M5S 1A8, Canada
| | - Mingkun Wu
- Donnelly Centre, University of Toronto, Toronto, M5S 3E1, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, M5S 1A8, Canada
| | - Zuyao Ni
- Donnelly Centre, University of Toronto, Toronto, M5S 3E1, Canada
| | - Hua Tang
- Donnelly Centre, University of Toronto, Toronto, M5S 3E1, Canada
| | - Guoqing Zhong
- Donnelly Centre, University of Toronto, Toronto, M5S 3E1, Canada
| | - Edyta Marcon
- Donnelly Centre, University of Toronto, Toronto, M5S 3E1, Canada
- Department of laboratory Medicine and Pathobiology, University of Toronto, Toronto, M5S 1A8, Canada
| | - Zhaolei Zhang
- Donnelly Centre, University of Toronto, Toronto, M5S 3E1, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, M5S 1A8, Canada
- Department of Computer Sciences, University of Toronto, Toronto, M5S 1A8, Canada
| | - Benjamin J Blencowe
- Donnelly Centre, University of Toronto, Toronto, M5S 3E1, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, M5S 1A8, Canada
| | - Jack F Greenblatt
- Donnelly Centre, University of Toronto, Toronto, M5S 3E1, Canada.
- Department of Molecular Genetics, University of Toronto, Toronto, M5S 1A8, Canada.
| |
Collapse
|
9
|
Bei Y, Zhu Y, Zhou J, Ai S, Yao J, Yin M, Hu M, Qi W, Spanos M, Li L, Wei M, Huang Z, Gao J, Liu C, van der Kraak PH, Li G, Lei Z, Sluijter JPG, Xiao J. Inhibition of Hmbox1 Promotes Cardiomyocyte Survival and Glucose Metabolism Through Gck Activation in Ischemia/Reperfusion Injury. Circulation 2024; 150:848-866. [PMID: 38708602 DOI: 10.1161/circulationaha.123.067592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Accepted: 04/11/2024] [Indexed: 05/07/2024]
Abstract
BACKGROUND Exercise-induced physiological cardiac growth regulators may protect the heart from ischemia/reperfusion (I/R) injury. Homeobox-containing 1 (Hmbox1), a homeobox family member, has been identified as a putative transcriptional repressor and is downregulated in the exercised heart. However, its roles in exercise-induced physiological cardiac growth and its potential protective effects against cardiac I/R injury remain largely unexplored. METHODS We studied the function of Hmbox1 in exercise-induced physiological cardiac growth in mice after 4 weeks of swimming exercise. Hmbox1 expression was then evaluated in human heart samples from deceased patients with myocardial infarction and in the animal cardiac I/R injury model. Its role in cardiac I/R injury was examined in mice with adeno-associated virus 9 (AAV9) vector-mediated Hmbox1 knockdown and in those with cardiac myocyte-specific Hmbox1 ablation. We performed RNA sequencing, promoter prediction, and binding assays and identified glucokinase (Gck) as a downstream effector of Hmbox1. The effects of Hmbox1 together with Gck were examined in cardiomyocytes to evaluate their cell size, proliferation, apoptosis, mitochondrial respiration, and glycolysis. The function of upstream regulator of Hmbox1, ETS1, was investigated through ETS1 overexpression in cardiac I/R mice in vivo. RESULTS We demonstrated that Hmbox1 downregulation was required for exercise-induced physiological cardiac growth. Inhibition of Hmbox1 increased cardiomyocyte size in isolated neonatal rat cardiomyocytes and human embryonic stem cell-derived cardiomyocytes but did not affect cardiomyocyte proliferation. Under pathological conditions, Hmbox1 was upregulated in both human and animal postinfarct cardiac tissues. Furthermore, both cardiac myocyte-specific Hmbox1 knockout and AAV9-mediated Hmbox1 knockdown protected against cardiac I/R injury and heart failure. Therapeutic effects were observed when sh-Hmbox1 AAV9 was administered after I/R injury. Inhibition of Hmbox1 activated the Akt/mTOR/P70S6K pathway and transcriptionally upregulated Gck, leading to reduced apoptosis and improved mitochondrial respiration and glycolysis in cardiomyocytes. ETS1 functioned as an upstream negative regulator of Hmbox1 transcription, and its overexpression was protective against cardiac I/R injury. CONCLUSIONS Our studies unravel a new role for the transcriptional repressor Hmbox1 in exercise-induced physiological cardiac growth. They also highlight the therapeutic potential of targeting Hmbox1 to improve myocardial survival and glucose metabolism after I/R injury.
Collapse
Affiliation(s)
- Yihua Bei
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital (Sixth People's Hospital of Nantong) and School of Life Science of Shanghai University, China (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.)
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education) (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China
- Cardiac Regeneration and Ageing Laboratory, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China
| | - Yujiao Zhu
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital (Sixth People's Hospital of Nantong) and School of Life Science of Shanghai University, China (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.)
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education) (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China
- Cardiac Regeneration and Ageing Laboratory, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China
| | - Jingwen Zhou
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital (Sixth People's Hospital of Nantong) and School of Life Science of Shanghai University, China (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.)
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education) (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China
- Cardiac Regeneration and Ageing Laboratory, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China
| | - Songwei Ai
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital (Sixth People's Hospital of Nantong) and School of Life Science of Shanghai University, China (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.)
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education) (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China
- Cardiac Regeneration and Ageing Laboratory, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China
| | - Jianhua Yao
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, China (J.Y.)
| | - Mingming Yin
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital (Sixth People's Hospital of Nantong) and School of Life Science of Shanghai University, China (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.)
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education) (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China
- Cardiac Regeneration and Ageing Laboratory, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China
| | - Meiyu Hu
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital (Sixth People's Hospital of Nantong) and School of Life Science of Shanghai University, China (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.)
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education) (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China
- Cardiac Regeneration and Ageing Laboratory, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China
| | - Weitong Qi
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital (Sixth People's Hospital of Nantong) and School of Life Science of Shanghai University, China (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.)
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education) (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China
- Cardiac Regeneration and Ageing Laboratory, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China
| | - Michail Spanos
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston (M.S., G.L.)
| | - Lin Li
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital (Sixth People's Hospital of Nantong) and School of Life Science of Shanghai University, China (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.)
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education) (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China
- Cardiac Regeneration and Ageing Laboratory, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China
| | - Meng Wei
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital (Sixth People's Hospital of Nantong) and School of Life Science of Shanghai University, China (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.)
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education) (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China
- Cardiac Regeneration and Ageing Laboratory, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China
| | - Zhenzhen Huang
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital (Sixth People's Hospital of Nantong) and School of Life Science of Shanghai University, China (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.)
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education) (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China
- Cardiac Regeneration and Ageing Laboratory, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China
| | - Juan Gao
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital (Sixth People's Hospital of Nantong) and School of Life Science of Shanghai University, China (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.)
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education) (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China
- Cardiac Regeneration and Ageing Laboratory, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China
| | - Chang Liu
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital (Sixth People's Hospital of Nantong) and School of Life Science of Shanghai University, China (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.)
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education) (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China
- Cardiac Regeneration and Ageing Laboratory, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China
| | - Petra H van der Kraak
- Department of Pathology (P.H.v.d.K.), University Medical Center Utrecht, University Utrecht, The Netherlands
| | - Guoping Li
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston (M.S., G.L.)
| | - Zhiyong Lei
- Department of Cardiology, Laboratory of Experimental Cardiology (Z.L., J.P.G.S.), University Medical Center Utrecht, University Utrecht, The Netherlands
- Division Laboratory, Central Diagnosis Laboratory Research (Z.L.), University Medical Center Utrecht, University Utrecht, The Netherlands
| | - Joost P G Sluijter
- Department of Cardiology, Laboratory of Experimental Cardiology (Z.L., J.P.G.S.), University Medical Center Utrecht, University Utrecht, The Netherlands
- Utrecht Regenerative Medicine Center (J.P.G.S.), University Medical Center Utrecht, University Utrecht, The Netherlands
| | - Junjie Xiao
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital (Sixth People's Hospital of Nantong) and School of Life Science of Shanghai University, China (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.)
- Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education) (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China
- Cardiac Regeneration and Ageing Laboratory, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine (Y.B., Y.Z., J.Z., S.A., M.Y., M.H., W.Q., L.L., M.W., Z.H., J.G., C.L., J.X.), Shanghai University, China
| |
Collapse
|
10
|
Yang Y, Gao F, Ren L, Ren N, Pan J, Xu Q. Functions and mechanisms of RNA m 6A regulators in breast cancer (Review). Int J Oncol 2024; 65:86. [PMID: 39054967 PMCID: PMC11299767 DOI: 10.3892/ijo.2024.5674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 07/09/2024] [Indexed: 07/27/2024] Open
Abstract
Breast cancer (BC) is a major malignant tumor in females and the incidence rate of BC has increased worldwide in recent years. N6‑methyladenosine (m6A) is a methylation modification that occurs extensively in eukaryotic RNA. The abnormal expression of m6A and related regulatory proteins can activate or inhibit certain signal pathways or oncogenes, thus affecting the proliferation, metastasis and prognosis of BC. Numerous studies have shown that m6A regulator disorder exists in BC, and this disorder can be reversed. Therefore, m6A is predicted as a potential therapeutic target for BC. However, the molecular mechanism of m6A RNA methylation regulating the occurrence and development of BC has not been comprehensively elucidated. In this review article, the functions of various m6A regulators and the specific mechanisms of certain regulators of the progress of BC were summarized. Furthermore, the dual role of RNA methylation in tumor progression was discussed, concluding that RNA methylation can not only lead to tumorigenesis but at times give rise to inhibition of tumor formation. In addition, further comprehensive analysis on mechanisms of m6A regulators in BC is conducive to screening effective potential targets and formulating targeted treatment strategies, which will provide new methods for the prevention and treatment of BC.
Collapse
Affiliation(s)
- Yibei Yang
- Department of Clinical Pharmacology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Cancer Center, Westlake University School of Medicine, Hangzhou, Zhejiang 310006, P.R. China
- Fourth Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310051, P.R. China
| | - Feng Gao
- Department of Urology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang 310000, P.R. China
| | - Lanqi Ren
- Fourth Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310051, P.R. China
| | - Ning Ren
- Fourth Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310051, P.R. China
| | - Junjie Pan
- Fourth Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310051, P.R. China
| | - Qiaoping Xu
- Department of Clinical Pharmacology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Cancer Center, Westlake University School of Medicine, Hangzhou, Zhejiang 310006, P.R. China
| |
Collapse
|
11
|
Han X, Zhu Y, Ke J, Zhai Y, Huang M, Zhang X, He H, Zhang X, Zhao X, Guo K, Li X, Han Z, Zhang Y. Progression of m 6A in the tumor microenvironment: hypoxia, immune and metabolic reprogramming. Cell Death Discov 2024; 10:331. [PMID: 39033180 PMCID: PMC11271487 DOI: 10.1038/s41420-024-02092-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 07/03/2024] [Accepted: 07/05/2024] [Indexed: 07/23/2024] Open
Abstract
Recently, N6-methyladenosine (m6A) has aroused widespread discussion in the scientific community as a mode of RNA modification. m6A comprises writers, erasers, and readers, which regulates RNA production, nuclear export, and translation and is very important for human health. A large number of studies have found that the regulation of m6A is closely related to the occurrence and invasion of tumors, while the homeostasis and function of the tumor microenvironment (TME) determine the occurrence and development of tumors to some extent. TME is composed of a variety of immune cells (T cells, B cells, etc.) and nonimmune cells (tumor-associated mesenchymal stem cells (TA-MSCs), cancer-associated fibroblasts (CAFs), etc.). Current studies suggest that m6A is involved in regulating the function of various cells in the TME, thereby affecting tumor progression. In this manuscript, we present the composition of m6A and TME, the relationship between m6A methylation and characteristic changes in TME, the role of m6A methylation in TME, and potential therapeutic strategies to provide new perspectives for better treatment of tumors in clinical work.
Collapse
Affiliation(s)
- Xuan Han
- First Clinical College of Changzhi Medical College, Changzhi, China
| | - Yu Zhu
- Linfen Central Hospital, Linfen, China
| | - Juan Ke
- Linfen Central Hospital, Linfen, China
| | | | - Min Huang
- Linfen Central Hospital, Linfen, China
| | - Xin Zhang
- Linfen Central Hospital, Linfen, China
| | | | | | | | | | | | - Zhongyu Han
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | | |
Collapse
|
12
|
Hara T, Meng S, Sato H, Tatekawa S, Sasaki K, Takeda Y, Tsuji Y, Arao Y, Ofusa K, Kitagawa T, Yamada D, Takahashi H, Kobayashi S, Motooka D, Suzuki Y, Rennie S, Uchida S, Mori M, Ogawa K, Doki Y, Eguchi H, Ishii H. High N6-methyladenosine-activated TCEAL8 mRNA is a novel pancreatic cancer marker. Cancer Sci 2024; 115:2360-2370. [PMID: 38659235 PMCID: PMC11247549 DOI: 10.1111/cas.16152] [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: 12/05/2023] [Revised: 02/22/2024] [Accepted: 03/03/2024] [Indexed: 04/26/2024] Open
Abstract
N6-methyladenosine (m6A) is an RNA modification involved in RNA processing and widely found in transcripts. In cancer cells, m6A is upregulated, contributing to their malignant transformation. In this study, we analyzed gene expression and m6A modification in cancer tissues, ducts, and acinar cells derived from pancreatic cancer patients using MeRIP-seq. We found that dozens of RNAs highly modified by m6A were detected in cancer tissues compared with ducts and acinar cells. Among them, the m6A-activated mRNA TCEAL8 was observed, for the first time, as a potential marker gene in pancreatic cancer. Spatially resolved transcriptomic analysis showed that TCEAL8 was highly expressed in specific cells, and activation of cancer-related signaling pathways was observed relative to TCEAL8-negative cells. Furthermore, among TCEAL8-positive cells, the cells expressing the m6A-modifying enzyme gene METTL3 showed co-activation of Notch and mTOR signaling, also known to be involved in cancer metastasis. Overall, these results suggest that m6A-activated TCEAL8 is a novel marker gene involved in the malignant transformation of pancreatic cancer.
Collapse
Affiliation(s)
- Tomoaki Hara
- Department of Medical Data Science, Center of Medical Innovation and Translational ResearchOsaka University Graduate School of MedicineSuitaOsakaJapan
| | - Sikun Meng
- Department of Medical Data Science, Center of Medical Innovation and Translational ResearchOsaka University Graduate School of MedicineSuitaOsakaJapan
| | - Hiromichi Sato
- Department of Medical Data Science, Center of Medical Innovation and Translational ResearchOsaka University Graduate School of MedicineSuitaOsakaJapan
- Department of Gastroenterological SurgeryOsaka University Graduate School of MedicineSuitaOsakaJapan
| | - Shotaro Tatekawa
- Department of Radiation OncologyOsaka University Graduate School of MedicineSuitaOsakaJapan
| | - Kazuki Sasaki
- Department of Medical Data Science, Center of Medical Innovation and Translational ResearchOsaka University Graduate School of MedicineSuitaOsakaJapan
- Department of Gastroenterological SurgeryOsaka University Graduate School of MedicineSuitaOsakaJapan
| | - Yu Takeda
- Department of Medical Data Science, Center of Medical Innovation and Translational ResearchOsaka University Graduate School of MedicineSuitaOsakaJapan
- Department of Gastroenterological SurgeryOsaka University Graduate School of MedicineSuitaOsakaJapan
| | - Yoshiko Tsuji
- Department of Medical Data Science, Center of Medical Innovation and Translational ResearchOsaka University Graduate School of MedicineSuitaOsakaJapan
| | - Yasuko Arao
- Department of Medical Data Science, Center of Medical Innovation and Translational ResearchOsaka University Graduate School of MedicineSuitaOsakaJapan
| | - Ken Ofusa
- Department of Medical Data Science, Center of Medical Innovation and Translational ResearchOsaka University Graduate School of MedicineSuitaOsakaJapan
- Prophoenix DivisionFood and Life‐Science Laboratory, IDEA Consultants, Inc.OsakaOsakaJapan
| | - Toru Kitagawa
- Department of Medical Data Science, Center of Medical Innovation and Translational ResearchOsaka University Graduate School of MedicineSuitaOsakaJapan
- Department of Gastroenterological SurgeryOsaka University Graduate School of MedicineSuitaOsakaJapan
- Kyowa‐kai Medical CorporationKawanishiHyogoJapan
| | - Daisaku Yamada
- Department of Gastroenterological SurgeryOsaka University Graduate School of MedicineSuitaOsakaJapan
| | - Hidenori Takahashi
- Department of Gastroenterological SurgeryOsaka University Graduate School of MedicineSuitaOsakaJapan
| | - Shogo Kobayashi
- Department of Gastroenterological SurgeryOsaka University Graduate School of MedicineSuitaOsakaJapan
| | - Daisuke Motooka
- Genome Information Research Center, Research Institute for Microbial DiseasesOsaka UniversitySuitaOsakaJapan
| | - Yutaka Suzuki
- Laboratory of Systems Genomics, Department of Computational Biology and Medical Sciences, Graduate School of Frontier SciencesThe University of TokyoKashiwa‐shiChibaJapan
| | - Sarah Rennie
- Section for Computational and RNA Biology, Department of BiologyUniversity of CopenhagenCopenhagenDenmark
| | - Shizuka Uchida
- Department of Clinical Medicine, Center for RNA MedicineAalborg UniversityCopenhagen SVDenmark
| | - Masaki Mori
- Tokai University Graduate School of MedicineIseharaKanagawaJapan
| | - Kazuhiko Ogawa
- Department of Radiation OncologyOsaka University Graduate School of MedicineSuitaOsakaJapan
| | - Yuichiro Doki
- Department of Gastroenterological SurgeryOsaka University Graduate School of MedicineSuitaOsakaJapan
| | - Hidetoshi Eguchi
- Department of Gastroenterological SurgeryOsaka University Graduate School of MedicineSuitaOsakaJapan
| | - Hideshi Ishii
- Department of Medical Data Science, Center of Medical Innovation and Translational ResearchOsaka University Graduate School of MedicineSuitaOsakaJapan
| |
Collapse
|
13
|
Zhang C, Yu M, Hepperla AJ, Zhang Z, Raj R, Zhong H, Zhou J, Hu L, Fang J, Liu H, Liang Q, Jia L, Liao C, Xi S, Simon JM, Xu K, Liu Z, Nam Y, Kapur P, Zhang Q. Von Hippel Lindau tumor suppressor controls m6A-dependent gene expression in renal tumorigenesis. J Clin Invest 2024; 134:e175703. [PMID: 38618952 PMCID: PMC11014668 DOI: 10.1172/jci175703] [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: 09/11/2023] [Accepted: 03/01/2024] [Indexed: 04/16/2024] Open
Abstract
N6-Methyladenosine (m6A) is the most abundant posttranscriptional modification, and its contribution to cancer evolution has recently been appreciated. Renal cancer is the most common adult genitourinary cancer, approximately 85% of which is accounted for by the clear cell renal cell carcinoma (ccRCC) subtype characterized by VHL loss. However, it is unclear whether VHL loss in ccRCC affects m6A patterns. In this study, we demonstrate that VHL binds and promotes METTL3/METTL14 complex formation while VHL depletion suppresses m6A modification, which is distinctive from its canonical E3 ligase role. m6A RNA immunoprecipitation sequencing (RIP-Seq) coupled with RNA-Seq allows us to identify a selection of genes whose expression may be regulated by VHL-m6A signaling. Specifically, PIK3R3 is identified to be a critical gene whose mRNA stability is regulated by VHL in a m6A-dependent but HIF-independent manner. Functionally, PIK3R3 depletion promotes renal cancer cell growth and orthotopic tumor growth while its overexpression leads to decreased tumorigenesis. Mechanistically, the VHL-m6A-regulated PIK3R3 suppresses tumor growth by restraining PI3K/AKT activity. Taken together, we propose a mechanism by which VHL regulates m6A through modulation of METTL3/METTL14 complex formation, thereby promoting PIK3R3 mRNA stability and protein levels that are critical for regulating ccRCC tumorigenesis.
Collapse
Affiliation(s)
- Cheng Zhang
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Miaomiao Yu
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Austin J. Hepperla
- Lineberger Comprehensive Cancer Center, University of North Carolina (UNC) School of Medicine, Chapel Hill, North Carolina, USA
- Department of Genetics, Neuroscience Center and
- UNC Neuroscience Center, Carolina Institute for Developmental Disabilities, UNC, Chapel Hill, North Carolina, USA
| | - Zhao Zhang
- Department of Molecular Medicine, Mays Cancer Center, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Rishi Raj
- Department of Biochemistry, Department of Biophysics, Simmons Comprehensive Cancer Center and
| | - Hua Zhong
- Department of Pathology, Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Jin Zhou
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Lianxin Hu
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Jun Fang
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Hongyi Liu
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Qian Liang
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Liwei Jia
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Chengheng Liao
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Sichuan Xi
- Thoracic Epigenetics Section, Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Jeremy M. Simon
- Lineberger Comprehensive Cancer Center, University of North Carolina (UNC) School of Medicine, Chapel Hill, North Carolina, USA
- Department of Genetics, Neuroscience Center and
- UNC Neuroscience Center, Carolina Institute for Developmental Disabilities, UNC, Chapel Hill, North Carolina, USA
| | - Kexin Xu
- Department of Molecular Medicine, Mays Cancer Center, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Zhijie Liu
- Department of Molecular Medicine, Mays Cancer Center, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Yunsun Nam
- Department of Biochemistry, Department of Biophysics, Simmons Comprehensive Cancer Center and
| | - Payal Kapur
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, Department of Urology
| | - Qing Zhang
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
14
|
Zhao C, Yu M, Li Y. Pan-cancer analysis reveals the pro-oncogenic role of N6-methyladenosine (m6A)-regulated NTMT1 in head and neck squamous cell carcinoma. J Biochem Mol Toxicol 2024; 38:e23603. [PMID: 38014887 DOI: 10.1002/jbt.23603] [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: 02/14/2023] [Revised: 09/09/2023] [Accepted: 11/20/2023] [Indexed: 11/29/2023]
Abstract
Head and neck squamous cell carcinoma (HNSC) is a common and fatal tumor with a bleak prognosis, posing a significant threat to human health. N6-methyladenosine (m6A) modification regulates tumor progression by modulating gene expression post-transcriptionally. Nevertheless, the specific function of m6A-modified tumor drivers in HNSC remains largely uncharted. In this study, we revealed the pro-oncogenic role of m6A-regulated NTMT1 in HNSC through comprehensive pan-cancer analysis and experimental validation. By scrutinizing the prognostic and expression profiles of NTMT1 across over 30 cancer types, we observed a significant association between NTMT1 and patient overall survival in ACC, HNSC, LAML, LGG, KIRC, and STAD. Moreover, we find a close correlation between NTMT1 and disease-free survival in ACC, HNSC, LUSC, UVM, KIRC, and STAD. NTMT1 exhibited dysregulation in 15 cancers, including CESC, CHOL, COAD, DLBC, GBM, HNSC, LGG, LIHC, PAAD, READ, SKCM, THYM, UCS, LAML, and TGCT. Integrated data underscored the critical involvement of NTMT1 in HNSC. Furthermore, the expression of NTMT1 was closely associated with tumor stage and immune infiltration in HNSC. Functionally, NTMT1 deficiency was demonstrated to significantly impede cell proliferation and cell-cycle progression in HNSC. Mechanistically, METTL3 was elucidated to mediate the epigenetic upregulation of NTMT1 in HNSC in an m6A-dependent manner, and the overexpression of METTL3 was shown to alleviate the inhibitory impact of downregulated NTMT1 on HNSC proliferation. In conclusion, our findings enhance our understanding of NTMT1's role across various cancer types and offer a rationale for clinically targeting NTMT1 as a therapeutic approach for HNSC.
Collapse
Affiliation(s)
- Chunhong Zhao
- Department of Otolaryngology-Head and Neck Surgery, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Min Yu
- Department of Otolaryngology-Head and Neck Surgery, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Yujie Li
- Department of Otolaryngology-Head and Neck Surgery, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| |
Collapse
|
15
|
Ochi S, Roy B, Prall K, Shelton RC, Dwivedi Y. Strong associations of telomere length and mitochondrial copy number with suicidality and abuse history in adolescent depressed individuals. Mol Psychiatry 2023; 28:3920-3929. [PMID: 37735501 PMCID: PMC10730407 DOI: 10.1038/s41380-023-02263-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 08/30/2023] [Accepted: 09/08/2023] [Indexed: 09/23/2023]
Abstract
Major depressive disorder (MDD) is highly prevalent in adolescents and is a major risk factor for suicidality. Recent evidence shows that accelerated cellular senescence/aging is associated with psychiatric illness, including depression, in adults. The present study examined if the relationships of telomere length (TL) and mitochondrial DNA copy number (mtDNAcn), two critical indicators of cellular senescence/aging, are altered in depressed adolescents and whether these alterations are associated with suicidality, early-life adversities, and other co-occuring factors. In genomic DNA isolated from 53 adolescents (ages 16-19, 19 MDD with suicide attempt/suicidal ideation [MDD + SI/SA], 14 MDD without SA/SI [MDD-SI/SA], and 20 healthy controls [HC]), TL and mtDNAcn were measured as the ratio between the number of telomere repeats and that of a single-copy nuclear-hemoglobin [HBG] gene or the amount of mtDNA (NADH dehydrogenase, subunit 1) relative to HBG. Our data show that TL was significantly lower, and mtDNAcn was significantly higher in the total MDD group than HC. TL was significantly lower and mtDNAcn was significantly higher in the MDD + SA/SI group than in the HC, whereas there were no differences in the MDD-SI/SA group. TL was positively correlated with mtDNAcn in both HC and MDD-SA/SI groups; however, TL was negatively correlated with mtDNAcn in MDD + SA/SI. Furthermore, TL was negatively correlated with the severity of both depression and anxiety, while mtDNAcn was positively correlated with the severity of prior emotional abuse. Our study indicates that cellular senescence is more advanced in depressed adolescents with suicidal ideation and that childhood emotional abuse may participate in such a process.
Collapse
Affiliation(s)
- Shinichiro Ochi
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
- Department of Neuropsychiatry, Molecules and Function, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime, 791-0295, Japan
| | - Bhaskar Roy
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Kevin Prall
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Richard C Shelton
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Yogesh Dwivedi
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
| |
Collapse
|
16
|
Liu R, Zhao E, Yu H, Yuan C, Abbas MN, Cui H. Methylation across the central dogma in health and diseases: new therapeutic strategies. Signal Transduct Target Ther 2023; 8:310. [PMID: 37620312 PMCID: PMC10449936 DOI: 10.1038/s41392-023-01528-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/23/2023] [Accepted: 05/25/2023] [Indexed: 08/26/2023] Open
Abstract
The proper transfer of genetic information from DNA to RNA to protein is essential for cell-fate control, development, and health. Methylation of DNA, RNAs, histones, and non-histone proteins is a reversible post-synthesis modification that finetunes gene expression and function in diverse physiological processes. Aberrant methylation caused by genetic mutations or environmental stimuli promotes various diseases and accelerates aging, necessitating the development of therapies to correct the disease-driver methylation imbalance. In this Review, we summarize the operating system of methylation across the central dogma, which includes writers, erasers, readers, and reader-independent outputs. We then discuss how dysregulation of the system contributes to neurological disorders, cancer, and aging. Current small-molecule compounds that target the modifiers show modest success in certain cancers. The methylome-wide action and lack of specificity lead to undesirable biological effects and cytotoxicity, limiting their therapeutic application, especially for diseases with a monogenic cause or different directions of methylation changes. Emerging tools capable of site-specific methylation manipulation hold great promise to solve this dilemma. With the refinement of delivery vehicles, these new tools are well positioned to advance the basic research and clinical translation of the methylation field.
Collapse
Affiliation(s)
- Ruochen Liu
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing, 400715, China
- Jinfeng Laboratory, Chongqing, 401329, China
- Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Chongqing, 400716, China
- Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, Chongqing, 400715, China
| | - Erhu Zhao
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing, 400715, China
- Jinfeng Laboratory, Chongqing, 401329, China
- Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Chongqing, 400716, China
- Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, Chongqing, 400715, China
| | - Huijuan Yu
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing, 400715, China
| | - Chaoyu Yuan
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing, 400715, China
| | - Muhammad Nadeem Abbas
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing, 400715, China
- Jinfeng Laboratory, Chongqing, 401329, China
- Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Chongqing, 400716, China
- Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, Chongqing, 400715, China
| | - Hongjuan Cui
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing, 400715, China.
- Jinfeng Laboratory, Chongqing, 401329, China.
- Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Chongqing, 400716, China.
- Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, Chongqing, 400715, China.
| |
Collapse
|
17
|
Tang F, Chen X, Liu JS, Liu ZY, Yang JZ, Wang ZF, Li ZQ. TERT mutations-associated alterations in clinical characteristics, immune environment and therapy response in glioblastomas. Discov Oncol 2023; 14:148. [PMID: 37566174 PMCID: PMC10421840 DOI: 10.1007/s12672-023-00760-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 07/25/2023] [Indexed: 08/12/2023] Open
Abstract
OBJECTIVE TERT: is the most frequently mutated gene in adult glioblastomas (GBMs) defined by the 2021 World Health Organization classification system. The present study aims to explore differences in clinical characteristics and immune microenvironment between TERT mutant and wild-type GBM. METHODS Three GBM-related cohorts consisting of 205 GBM patients in our cohort, 463 GBM patients without immune checkpoint inhibitor(ICI) therapy and 1465 tumour patients (including 92 GBM cases) receiving ICI treatment in the MSK cohort were included. Retrospective analysis and immunohistochemistry assay were used for investigating the local (including tumour cells, local immune cells, and seizures) and systemic (including circulating immune cells, coagulation-related functions, and prognosis) effects of TERT mutations. Besides, differences in genetic alterations and immunotherapy responses between TERT mutant and wild-type GBMs were also explored. RESULTS We found that TERT mutant and wild-type GBMs possessed similar initial clinic symptoms, circulating immune microenvironment and immunotherapy response. With respect to that in TERT wild-type GBMs, mutations in TERT resulted in higher levels of tumour-infiltrating neutrophils, prolonged coagulation time, worse chemotherapy response and poorer overall survival. CONCLUSION Mutations in TERT alter the local immune environment and decrease the sensitivity of GBM to chemotherapy.
Collapse
Affiliation(s)
- Feng Tang
- Brain Glioma Center, Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Xi Chen
- Brain Glioma Center, Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Jin-Sheng Liu
- Brain Glioma Center, Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Zhen-Yuan Liu
- Brain Glioma Center, Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Jin-Zhou Yang
- Brain Glioma Center, Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Ze-Fen Wang
- Department of Physiology, Wuhan University School of Basic Medical Sciences, Wuhan, Hubei, China.
| | - Zhi-Qiang Li
- Brain Glioma Center, Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
18
|
Yi M, Wang M, Xu Y, Cao Z, Ling Y, Zhang Z, Cao H. CRISPR-based m 6A modification and its potential applications in telomerase regulation. Front Cell Dev Biol 2023; 11:1200734. [PMID: 37519297 PMCID: PMC10382234 DOI: 10.3389/fcell.2023.1200734] [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: 04/05/2023] [Accepted: 06/27/2023] [Indexed: 08/01/2023] Open
Abstract
Telomerase determines cell lifespan by controlling chromosome stability and cell viability, m6A epigenetic modification plays an important role in the regulation of telomerase activity. Using CRISPR epigenome editing to analyze specific m6A modification sites in telomerase will provide an important tool for analyzing the molecular mechanism of m6A modification regulating telomerase activity. In this review, we clarified the relevant applications of CRISPR system, paid special attention to the regulation of m6A modification in stem cells and cancer cells based on CRISPR system, emphasized the regulation of m6A modification on telomerase activity, pointed out that m6A modification sites regulate telomerase activity, and discussed strategies based on telomerase activity and disease treatment, which are helpful to promote the research of anti-aging and tumor related diseases.
Collapse
Affiliation(s)
- Mingliang Yi
- Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio-breeding, Anhui Agricultural University, Hefei, China
| | - Mingyue Wang
- Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio-breeding, Anhui Agricultural University, Hefei, China
| | - Yongjie Xu
- Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio-breeding, Anhui Agricultural University, Hefei, China
| | - Zhikun Cao
- Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio-breeding, Anhui Agricultural University, Hefei, China
| | - Yinghui Ling
- Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio-breeding, Anhui Agricultural University, Hefei, China
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Zijun Zhang
- Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio-breeding, Anhui Agricultural University, Hefei, China
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Hongguo Cao
- Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio-breeding, Anhui Agricultural University, Hefei, China
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| |
Collapse
|
19
|
Gao P, Yao F, Pang J, Yin K, Zhu X. m 6A methylation in cellular senescence of age-associated diseases. Acta Biochim Biophys Sin (Shanghai) 2023; 55:1168-1183. [PMID: 37394885 PMCID: PMC10449638 DOI: 10.3724/abbs.2023107] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 04/14/2023] [Indexed: 07/04/2023] Open
Abstract
Cellular senescence is a state of irreversible cellular growth arrest that occurs in response to various stresses. In addition to exiting the cell cycle, senescent cells undergo many phenotypic alterations, including metabolic reprogramming, chromatin rearrangement, and senescence-associated secretory phenotype (SASP) development. Furthermore, senescent cells can affect most physiological and pathological processes, such as physiological development; tissue homeostasis; tumour regression; and age-associated disease progression, including diabetes, atherosclerosis, Alzheimer's disease, and hypertension. Although corresponding anti-senescence therapies are actively being explored for the treatment of age-associated diseases, the specific regulatory mechanisms of senescence remain unclear. N 6-methyladenosine (m 6A), a chemical modification commonly distributed in eukaryotic RNA, plays an important role in biological processes such as translation, shearing, and RNA transcription. Numerous studies have shown that m 6A plays an important regulatory role in cellular senescence and aging-related disease. In this review, we systematically summarize the role of m 6A modifications in cellular senescence with regard to oxidative stress, DNA damage, telomere alterations, and SASP development. Additionally, diabetes, atherosclerosis, and Alzheimer's disease regulation via m 6A-mediated cellular senescence is discussed. We further discuss the challenges and prospects of m 6A in cellular senescence and age-associated diseases with the aim of providing rational strategies for the treatment of these age-associated diseases.
Collapse
Affiliation(s)
- Pan Gao
- Guangxi Key Laboratory of Diabetic Systems MedicineGuilin Medical UniversityGuilin541100China
| | - Feng Yao
- Guangxi Key Laboratory of Diabetic Systems MedicineGuilin Medical UniversityGuilin541100China
| | - Jin Pang
- Guangxi Key Laboratory of Diabetic Systems MedicineGuilin Medical UniversityGuilin541100China
| | - Kai Yin
- The Fifth Affiliated Hospital of Southern Medical UniversityGuangzhou510900China
| | - Xiao Zhu
- Guangxi Key Laboratory of Diabetic Systems MedicineGuilin Medical UniversityGuilin541100China
| |
Collapse
|
20
|
Fu H, Zhu X, Di Q, Sun J, Jiang Q, Xu Q. m6A contributes to a pro-survival state in GC-2 cells by facilitating DNA damage repair: Novel perspectives on the mechanism underlying DEHP genotoxicity in male germ cells. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 859:160432. [PMID: 36423848 DOI: 10.1016/j.scitotenv.2022.160432] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/18/2022] [Accepted: 11/19/2022] [Indexed: 06/16/2023]
Abstract
Di(2-ethyl-hexyl) phthalate (DEHP), an environmental endocrine disruptor, can destroy the sperm genomic integrity and impairs spermatogenesis. N6-methyladenosine (m6A) is involved in the cellular effects of DEHP. However, the genotoxic effect of DEHP on spermatocytes and the possible role of m6A in this process remain unclear. This study demonstrated that m6A alleviates DEHP genotoxicity in GC-2 cells. In DEHP-treated mice, DNA double-strand breaks (DSBs) were induced in the testis and spermatocytes. To further explore the molecular mechanism of DEHP genotoxicity on spermatocytes, GC-2 cells were exposed to DEHP. DEHP produced distinct genotoxicity and caused DSBs, which led to the inhibition of DNA synthesis and cell cycle arrest. The DNA damage response (DDR) was initiated to repair the DSBs induced by environmentally relevant levels of DEHP (100 μM and 200 μM). During this process, METTL3 upregulated m6A, which facilitated the DDR via stabilizing the DNA damage repair factors (Rad51 and Xrcc5) mRNA to maintain the pro-survival state. Moreover, Mettl3 knockdown partially inhibited DDR. Interestingly, high-dose DEHP (400 μM and 600 μM) directly induced apoptosis rather than the pro-survival state. Altogether: METTL3-mediated m6A participates in maintaining the pro-survival state by upregulating DDR, providing guidance for mitigating the genotoxicity of environment-related level DEHP exposure.
Collapse
Affiliation(s)
- Haowei Fu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Xiaofang Zhu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Qiannan Di
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Jiahui Sun
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Qianqian Jiang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Qian Xu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China.
| |
Collapse
|
21
|
Orsolic I, Carrier A, Esteller M. Genetic and epigenetic defects of the RNA modification machinery in cancer. Trends Genet 2023; 39:74-88. [PMID: 36379743 DOI: 10.1016/j.tig.2022.10.004] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 09/25/2022] [Accepted: 10/26/2022] [Indexed: 11/13/2022]
Abstract
Cancer was initially considered to be an exclusively genetic disease, but an interplay of dysregulated genetic and epigenetic mechanisms is now known to contribute to the cancer phenotype. More recently, chemical modifications of RNA molecules - the so-called epitranscriptome - have been found to regulate various aspects of RNA function and homeostasis. Specific enzymes, known as RNA-modifying proteins (RMPs), are responsible for depositing, removing, and reading chemical modifications in RNA. Intensive investigations in the epitranscriptomic field in recent years, in conjunction with great technological advances, have revealed the critical role of RNA modifications in regulating numerous cellular pathways. Furthermore, growing evidence has revealed that RNA modification machinery is often altered in human cancers, highlighting the enormous potential of RMPs as pharmacological targets or diagnostic markers.
Collapse
Affiliation(s)
- Ines Orsolic
- Josep Carreras Leukemia Research Institute (IJC), Badalona, Barcelona, Catalonia, Spain
| | - Arnaud Carrier
- Josep Carreras Leukemia Research Institute (IJC), Badalona, Barcelona, Catalonia, Spain
| | - Manel Esteller
- Josep Carreras Leukemia Research Institute (IJC), Badalona, Barcelona, Catalonia, Spain; Centro de Investigacion Biomedica en Red Cancer (CIBERONC), 28029 Madrid, Spain; Institucio Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Catalonia, Spain; Physiological Sciences Department, School of Medicine and Health Sciences, University of Barcelona (UB), Barcelona, Catalonia, Spain.
| |
Collapse
|
22
|
Hong J, Xu K, Lee JH. Biological roles of the RNA m 6A modification and its implications in cancer. Exp Mol Med 2022; 54:1822-1832. [PMID: 36446846 PMCID: PMC9722703 DOI: 10.1038/s12276-022-00897-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 10/06/2022] [Accepted: 10/09/2022] [Indexed: 11/30/2022] Open
Abstract
The N6-Methyladenosine (m6A) modification of RNA transcripts is the most prevalent and abundant internal modification in eukaryotic messenger RNAs (mRNAs) and plays diverse and important roles in normal biological processes. Extensive studies have indicated that dysregulated m6A modification and m6A-associated proteins play critical roles in tumorigenesis and cancer progression. However, m6A-mediated physiological consequences often lead to opposite outcomes in a biological context-dependent manner. Therefore, context-related complexity must be meaningfully considered to obtain a comprehensive understanding of RNA methylation. Recently, it has been reported that m6A-modified RNAs are closely related to the regulation of the DNA damage response and genomic integrity maintenance. Here, we present an overview of the current knowledge on the m6A modification and its function in human cancer, particularly in relation to the DNA damage response and genomic instability.
Collapse
Affiliation(s)
- Juyeong Hong
- grid.267309.90000 0001 0629 5880Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX USA
| | - Kexin Xu
- grid.267309.90000 0001 0629 5880Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX USA
| | - Ji Hoon Lee
- grid.267309.90000 0001 0629 5880Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX USA
| |
Collapse
|
23
|
Zhu X, Fu H, Sun J, Di Q, Xu Q. N6-methyladenosine modification on Hmbox1 is related to telomere dysfunction in DEHP-induced male reproductive injury. Life Sci 2022; 309:121005. [PMID: 36174712 DOI: 10.1016/j.lfs.2022.121005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 09/12/2022] [Accepted: 09/23/2022] [Indexed: 11/19/2022]
Abstract
AIMS Di (2-ethylhexyl) phthalate (DEHP), as an environmental endocrine-disrupting chemical (EDC), can induce male reproductive injury. N6-methyladenosine (m6A) plays a vital role in environmental exposure-induced diseases by regulating gene expression. Therefore, we aim to investigate the role of m6A in DEHP-induced reproductive injury. MAIN METHODS We established an in vivo model of mice exposed to DEHP to explore the effect of DEHP on reproductive injury and m6A. To further explore the molecular mechanism of DEHP toxicity, we built a model of GC-2 cells exposed to mono-(2-ethylhexyl) phthalate (MEHP) in vitro and further silenced Mettl3 in GC-2cells. Besides, we also conducted MeRIP-qPCR and RIP assays to identify the target genes for m6A modification. KEY FINDINGS DEHP induced testicular injury and senescence. And telomeres shortening, reduced levels of telomere repeat-binding factor 1 (TRF1), TRF2, protection of telomeres 1 (POT1), and telomerase reverse transcriptase (TERT) can be observed in DEHP-treated testes. MEHP also induced GC-2 cellular senescence and telomere dysfunction. Besides, increased m6A mediated by METTL3 stabilized homeobox containing 1 (Hmbox1) in an m6A-dependent manner in MEHP-exposed GC-2 cells. Mettl3 knockdown led to lower m6A modification and reduced Hmbox1 stability, resulting in further shortening of telomere length. SIGNIFICANCE our work uncovered that DEHP led to male reproductive injury by telomere dysfunction and m6A modified Hmbox1 contributed to maintaining telomere homeostasis in this process, suggesting that accurate regulation of m6A modification level by drugs has potential value in the treatment of DEHP-induced male reproductive injury.
Collapse
Affiliation(s)
- Xiaofang Zhu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Haowei Fu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Jiahui Sun
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Qiannan Di
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Qian Xu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China.
| |
Collapse
|
24
|
Upregulation of PARG in prostate cancer cells suppresses their malignant behavior and downregulates tumor-promoting genes. Biomed Pharmacother 2022; 153:113504. [PMID: 36076593 DOI: 10.1016/j.biopha.2022.113504] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/24/2022] [Accepted: 07/30/2022] [Indexed: 02/03/2023] Open
Abstract
Post-translational modification of nuclear proteins through the addition of poly(ADP-ribose) (pADPr) moieties is upregulated in many metastatic cancers, where the high levels of pADPr have often been associated with poor cancer prognosis. Although the inhibitors of poly(ADP-ribose) polymerases (PARPs) have been utilized as potent anti-cancer agents, their efficacy in clinical trials varied among patient groups and has often been unpredictable. Such outcome cannot be interpreted solely by the inability to keep PARP-driven DNA repair in check. The focus of studies on PARP-driven tumorigenesis have recently been shifted toward PARP-dependent regulation of transcription. Here we utilized the controlled overexpression of poly(ADP-ribose) glycohydrolase (PARG), a sole pADPr-degrading enzyme, to investigate pADPr-dependent gene regulation in prostate cancer PC-3 cells. We demonstrated that PARG upregulation reduces pADPr levels and inhibits the expression of genes in key tumor-promoted pathways, including TNFα/NF-kB, IL6/STAT3, MYC, and KRAS signaling, the genes involved in inflammation response, especially chemokines, and endothelial-mesenchymal transition. The observed effect of PARG on transcription was consistent across all tested prostate cancer cell lines and correlates with PARG-induced reduction of clonogenic potential of PC-3 cells in vitro and a significant growth inhibition of PC-3-derived tumors in nude mice in vivo.
Collapse
|
25
|
Cai Z, Xu H, Bai G, Hu H, Wang D, Li H, Wang Z. ELAVL1 promotes prostate cancer progression by interacting with other m6A regulators. Front Oncol 2022; 12:939784. [PMID: 35978821 PMCID: PMC9376624 DOI: 10.3389/fonc.2022.939784] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 06/30/2022] [Indexed: 11/17/2022] Open
Abstract
N6-Methyladenosine (m6A) imbalance is an important factor in the occurrence and development of prostate cancer (PCa). Many m6A regulators have been found to be significantly dysregulated in PCa. ELAVL1 is an m6A binding protein that can promote the occurrence and development of tumors in an m6A-dependent manner. In this study, we found that most m6A regulators were significantly dysregulated in PCa, and some m6A regulators were associated with the progression-free interval. Mutations and copy number variations of these m6A regulators can alter their expression. However, ELAVL1 mutations were not found in PCa. Nevertheless, ELAVL1 upregulation was closely related to PCa proliferation. High ELAVL1 expression was also related to RNA metabolism. Further experiments showed that ELAVL1 interacted with other m6A regulators and that several m6A regulatory mRNAs have m6A sites that can be recognized by ELAVL1. Additionally, protein–protein interactions occur between ELAVL1 and other m6A regulators. Finally, we found that the dysregulation of ELAVL1 expression occurred in almost all tumors, and interactions between ELAVL1 and other m6A regulators also existed in almost all tumors. In summary, ELAVL1 is an important molecule in the development of PCa, and its interactions with other m6A regulators may play important roles in PCa progression.
Collapse
Affiliation(s)
- Zhonglin Cai
- Department of Urology, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Huan Xu
- Department of Urology, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Gang Bai
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Hanjing Hu
- Department of Biochemistry and Molecular Biology, School of Medicine, Nantong University, Nantong, China
| | - Di Wang
- Department of Molecular Pathology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China
- *Correspondence: Zhong Wang, ; Di Wang, ; Hongjun Li,
| | - Hongjun Li
- Department of Urology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
- *Correspondence: Zhong Wang, ; Di Wang, ; Hongjun Li,
| | - Zhong Wang
- Department of Urology, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- *Correspondence: Zhong Wang, ; Di Wang, ; Hongjun Li,
| |
Collapse
|
26
|
Zhang F, Liu H, Duan M, Wang G, Zhang Z, Wang Y, Qian Y, Yang Z, Jiang X. Crosstalk among m6A RNA methylation, hypoxia and metabolic reprogramming in TME: from immunosuppressive microenvironment to clinical application. J Hematol Oncol 2022; 15:84. [PMID: 35794625 PMCID: PMC9258089 DOI: 10.1186/s13045-022-01304-5] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 06/09/2022] [Indexed: 12/13/2022] Open
Abstract
The tumor microenvironment (TME), which is regulated by intrinsic oncogenic mechanisms and epigenetic modifications, has become a research hotspot in recent years. Characteristic features of TME include hypoxia, metabolic dysregulation, and immunosuppression. One of the most common RNA modifications, N6-methyladenosine (m6A) methylation, is widely involved in the regulation of physiological and pathological processes, including tumor development. Compelling evidence indicates that m6A methylation regulates transcription and protein expression through shearing, export, translation, and processing, thereby participating in the dynamic evolution of TME. Specifically, m6A methylation-mediated adaptation to hypoxia, metabolic dysregulation, and phenotypic shift of immune cells synergistically promote the formation of an immunosuppressive TME that supports tumor proliferation and metastasis. In this review, we have focused on the involvement of m6A methylation in the dynamic evolution of tumor-adaptive TME and described the detailed mechanisms linking m6A methylation to change in tumor cell biological functions. In view of the collective data, we advocate treating TME as a complete ecosystem in which components crosstalk with each other to synergistically achieve tumor adaptive changes. Finally, we describe the potential utility of m6A methylation-targeted therapies and tumor immunotherapy in clinical applications and the challenges faced, with the aim of advancing m6A methylation research.
Collapse
|
27
|
Lo N, Xu X, Soares F, He HH. The Basis and Promise of Programmable RNA Editing and Modification. Front Genet 2022; 13:834413. [PMID: 35154288 PMCID: PMC8831800 DOI: 10.3389/fgene.2022.834413] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 01/07/2022] [Indexed: 12/14/2022] Open
Abstract
One key advantage of RNA over genomic editing is its temporary effects. Aside from current use of DNA-targeting CRISPR-Cas9, the more recently discovered CRISPR-Cas13 has been explored as a means of editing due to its RNA-targeting capabilities. Specifically, there has been a recent interest in identifying and functionally characterizing biochemical RNA modifications, which has spurred a new field of research known as "epitranscriptomics". As one of the most frequently occurring transcriptome modifications, N6-methyladenosine (m6A) has generated much interest. The presence of m6A modifications is under the tight control of a series of regulators, and the ability of fusing these proteins or demethylases to catalytically inactive CRISPR proteins have resulted in a new wave of programmable RNA methylation tools. In addition, studies have been conducted to develop different CRISPR/Cas and base editor systems capable of more efficient editing, and some have explored the effects of in vivo editing for certain diseases. As well, the application of CRISPR and base editors for screening shows promise in revealing the phenotypic outcomes from m6A modification, many of which are linked to physiological, and pathological effects. Thus, the therapeutic potential of CRISPR/Cas and base editors for not only m6A related, but other RNA and DNA related disease has also garnered insight. In this review, we summarize/discuss the recent findings on RNA editing with CRISPR, base editors and non-CRISPR related tools and offer a perspective regarding future applications for basic and clinical research.
Collapse
Affiliation(s)
- Nicholas Lo
- Princess Margaret Cancer Center, University Health Network, Toronto, ON, Canada
| | - Xin Xu
- Princess Margaret Cancer Center, University Health Network, Toronto, ON, Canada
| | - Fraser Soares
- Princess Margaret Cancer Center, University Health Network, Toronto, ON, Canada
| | - Housheng Hansen He
- Princess Margaret Cancer Center, University Health Network, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| |
Collapse
|