1
|
Zhang Y, Cao X, Li W, Cui Z, Mao J, Yao R, Liu L. ALKBH5 reverses romidepsin-mediated anti-multiple myeloma activity via regulation of m6A modification of FOXM1. Biochem Pharmacol 2025; 239:116998. [PMID: 40419197 DOI: 10.1016/j.bcp.2025.116998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 04/07/2025] [Accepted: 05/22/2025] [Indexed: 05/28/2025]
Abstract
Multiple myeloma (MM), a prevalent hematological malignancy, is characterized by the abnormal proliferation and accumulation of plasma cells within the bone marrow. ALKBH5, an RNA demethylase, is implicated in various tumor-related processes, yet its role in myeloma remains insufficiently understood. In this study, we revealed that elevated ALKBH5 expression correlates with poor MM prognosis. Furthermore, romidepsin, a recognized histone deacetylase (HDAC) inhibitor, was found to significantly suppress both the protein levels and enzymatic activity of ALKBH5. Interestingly, while ectopic expression of ALKBH5 did not directly influence cell viability, its overexpression mitigated romidepsin-induced apoptosis. Conversely, ALKBH5 knockout enhanced the apoptotic effects of romidepsin, highlighting a potential synergistic interaction. Our findings also demonstrated that ALKBH5 attenuates romidepsin-mediated DNA damage through γH2AX activation. At the mechanistic level, ALKBH5 overexpression reduced m6A methylation of FOXM1 mRNA, stabilizing the transcript and promoting its translation. In contrast, romidepsin increased m6A methylation, accelerating FOXM1 mRNA decay. Notably, reintroduction of ALKBH5 reversed romidepsin's impact on FOXM1 expression. Additionally, CRISPR/Cas9-mediated ALKBH5 knockout enhanced the synergistic effects of romidepsin by inducing m6A-dependent FOXM1 mRNA destabilization. Our xenograft model further validated that ALKBH5 counteracts romidepsin-induced anti-MM activity by enhancing FOXM1 protein translation. Collectively, these findings illustrate that romidepsin exerts anti-MM effects by impairing ALKBH5 enzymatic activity and promoting FOXM1 mRNA m6A methylation. Therapeutic strategies combining ALKBH5 knockout with romidepsin may offer promising avenues for improving MM treatment outcomes.
Collapse
Affiliation(s)
- Yaxin Zhang
- Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xu Cao
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China; Department of Oncology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Wenjing Li
- Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Zeyu Cui
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jiwei Mao
- Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Ruosi Yao
- Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Linlin Liu
- College of Medical Imaging, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| |
Collapse
|
2
|
Liu X, Zeng S, Meng J, Zhou Q, Cao F, Chu B, Wu C, Wang Y, Feng H, Bi X, Chen X, Yang W, Tian M, Yang H, Hu K, Hou S. BARD1-mediated stabilization of METTL14 promotes retinal neovascularization by m6A-modifying MXD1 mRNA on a YTHDF2-dependent manner. Theranostics 2025; 15:5481-5498. [PMID: 40303324 PMCID: PMC12036877 DOI: 10.7150/thno.110122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 04/04/2025] [Indexed: 05/02/2025] Open
Abstract
Retinal vascular diseases are typified by the proliferation of irregular and leaky microvessels, resulting in vision impairment. Although the etiology of retinal angiogenesis is not yet fully understood, it is evident that microglia play a pivotal role in promoting angiogenesis. Methods: In vivo, the METTL14 conditional knockout (cKO) mouse was constructed to investigate the role of METTL14 in oxygen-induced retinopathy (OIR). In vitro, a combination of methylated RNA immunoprecipitation sequencing (MeRIP-seq), RNA-sequencing (RNA-seq), RNA Immunoprecipitation (RIP) assay, dual-luciferase reporter assays, and Chromatin immunoprecipitation-qPCR (ChIP-qPCR), was performed to explore the underlying mechanisms. Results: The proteomic analysis of hypoxic microglia has uncovered a pronounced enrichment in pathways related to RNA modification. Western blot has revealed that N6-methyladenosine (m6A) methyltransferase-like 14 (METTL14) exhibits the most significant increase among the RNA methylases. METTL14 cKO mice within an OIR model showed fewer neovascular formations. Additionally, in co-culture with sh-METTL14 HMC3 cells, HRMECs also exhibited reduced angiogenesis capabilities. Mechanically, E3 ubiquitin-protein ligase BARD1 can directly interact with METTL14, leading to an up-regulation of METTL14 protein level in hypoxic microglia. METTL14 could directly modifies and regulates the transcription factor MAX Dimerization Protein 1 (MXD1), which is subsequently recognized by the m6A "reader" YTH domain-containing family protein 2 (YTHDF2). Consequently, the modified MXD1 modulates the expression of VEGFA and VCAM1, promotes retinal neovascularization. Conclusion: Our study highlights the critical role of METTL14 in the OIR model and suggests a novel therapeutic target for addressing retinal vascular diseases.
Collapse
Affiliation(s)
- Xianyang Liu
- The First Affiliated Hospital of Chongqing Medical University, China
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing, 100730, China
| | - Shuhao Zeng
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing, 100730, China
| | - Jiayu Meng
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Qian Zhou
- The First Affiliated Hospital of Chongqing Medical University, China
| | - Fan Cao
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing, 100730, China
| | - Baorui Chu
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing, 100730, China
- Department of Ophthalmology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chao Wu
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing, 100730, China
| | - Yakun Wang
- The First Affiliated Hospital of Chongqing Medical University, China
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing, 100730, China
| | - Hui Feng
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing, 100730, China
| | - Xiaorui Bi
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing, 100730, China
| | - Xinyuan Chen
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing, 100730, China
| | - Wenxian Yang
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing, 100730, China
| | - Meng Tian
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing, 100730, China
| | - Hui Yang
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing, 100730, China
| | - Ke Hu
- The First Affiliated Hospital of Chongqing Medical University, China
| | - Shengping Hou
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing, 100730, China
| |
Collapse
|
3
|
Kapadia B, Roychowdhury A, Kayastha F, Lee WS, Nanaji N, Windle J, Gartenhaus R. m6A eraser ALKBH5/treRNA1/DDX46 axis regulates BCR expression. Neoplasia 2025; 62:101144. [PMID: 39987653 PMCID: PMC11905846 DOI: 10.1016/j.neo.2025.101144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 02/10/2025] [Accepted: 02/18/2025] [Indexed: 02/25/2025]
Abstract
Epitranscriptomic modifications, particularly N6-methyladenosine (m6A), have emerged as critical regulators of RNA stability, localization, and translation, shaping immune responses and tumor progression. In B-cell biology, m6A modifications influence germinal center formation and antigen-driven differentiation, underscoring their importance in immune regulation. Among m6A regulators, ALKBH5 (RNA demethylase) is pivotal in removing methylation marks and modulating gene expression in diverse cellular contexts. Despite advancements in understanding m6A dynamics, the mechanistic interplay between m6A demethylation and B-cell receptor (BCR) signaling pathways still needs to be explored. This study reveals a novel regulatory axis involving ALKBH5, treRNA1 (Translation Regulatory Long Non-Coding RNA 1), and DDX46 (RNA helicase). Upon activation signals, ALKBH5 and treRNA1 translocate to the nucleus, forming a functional complex with DDX46 to orchestrate the removal of m6A modifications on key transcripts, including those involved in BCR signaling. This demethylation enhances transcript stability and facilitates cytoplasmic export through interaction with the RNA-binding protein HuR, promoting efficient translation. Disruption of this axis, via loss of ALKBH5, DDX46, or treRNA1, led to impaired transcript processing and diminished BCR-related gene expression, highlighting the critical role of m6A demethylation in maintaining RNA dynamics. These findings uncover a previously unrecognized epitranscriptomic mechanism driven by the ALKBH5-treRNA1-DDX46 complex, with significant implications for B-cell functionality, immune regulation, and oncogenic pathways. Targeting this axis offers a promising avenue for developing therapeutic strategies in cancer and immune-related disorders where m6A dysregulation plays a central role.
Collapse
Affiliation(s)
- Bandish Kapadia
- Division of Hematology, Oncology, and Palliative Care, Department of Internal Medicine, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA; Section of Hematology and Oncology, Medicine Service, Richmond VA Cancer Center, Richmond Veteran Affairs Medical Center, Richmond, VA, USA; VCU Massey Comprehensive Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA 23298, USA.
| | - Anirban Roychowdhury
- Division of Hematology, Oncology, and Palliative Care, Department of Internal Medicine, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA; Section of Hematology and Oncology, Medicine Service, Richmond VA Cancer Center, Richmond Veteran Affairs Medical Center, Richmond, VA, USA; VCU Massey Comprehensive Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA 23298, USA
| | - Forum Kayastha
- Division of Hematology, Oncology, and Palliative Care, Department of Internal Medicine, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA; Section of Hematology and Oncology, Medicine Service, Richmond VA Cancer Center, Richmond Veteran Affairs Medical Center, Richmond, VA, USA; VCU Massey Comprehensive Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA 23298, USA
| | - Won Sok Lee
- Department of Pathology, Richmond Veteran Affairs Medical Center, Richmond, VA, USA
| | - Nahid Nanaji
- Department of Veteran Affairs, Maryland Healthcare System, Baltimore, MD, USA
| | - Jolene Windle
- VCU Massey Comprehensive Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA 23298, USA; Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA 23298, USA; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA 23298, USA
| | - Ronald Gartenhaus
- Division of Hematology, Oncology, and Palliative Care, Department of Internal Medicine, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA; Section of Hematology and Oncology, Medicine Service, Richmond VA Cancer Center, Richmond Veteran Affairs Medical Center, Richmond, VA, USA; VCU Massey Comprehensive Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA 23298, USA.
| |
Collapse
|
4
|
Cosseddu C, Succu S, Frau A, Mossa F, Versace SV, Brevini TAL, Ledda S, Bebbere D. m6A RNA methylation dynamics during in vitro maturation of cumulus-oocyte complexes derived from adult or prepubertal sheep. J Assist Reprod Genet 2025:10.1007/s10815-025-03444-2. [PMID: 40097858 DOI: 10.1007/s10815-025-03444-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 02/28/2025] [Indexed: 03/19/2025] Open
Abstract
PURPOSE N6-methyladenosine (m6A) is the most prevalent base epigenetic modification within eukaryotic mRNAs. It participates in post-transcriptional regulation, including maternal RNA maintenance and decay in mouse oocytes and during maternal-to-zygotic transition. The landscape in other mammalian species remains largely unexplored. The present work analyzed m6A dynamics in sheep cumulus oocyte complexes (COCs), during in vitro maturation. To explore potential relationships with oocyte developmental competence, a previously established model consisting of oocytes derived from adult and prepubertal sheep was adopted. METHODS m6a dynamics were analyzed in terms of m6A RNA methylation abundance in cumulus cells (CCs) by colorimetric assay and expression of key m6A methylation-related proteins (METTL3, METTL14, METTL16, VIRMA, YTHDC1, YTHDC2, YTHDF2, YTHDF3, ALKBH5, and FTO) in both cumulus cells and oocytes by real-time PCR. RESULTS We report the dynamics of m6A in sheep COCs, and reveal alterations in both oocytes and cumulus cells derived from prepubertal donors. These changes were observed in terms of m6A RNA methylation levels and transcript dynamics of several m6A methylation-related proteins. Notably, our study shows that dysregulations occur after IVM. CONCLUSION Overall, this work describes for the first time the dynamics of m6A in sheep COCs and uncovers the involvement of m6A RNA methylation in oocyte developmental potential.
Collapse
Affiliation(s)
- Chiara Cosseddu
- Department of Veterinary Medicine, Obstetrics and Gynecology Clinics, University of Sassari, 07100, Sassari, Italy
| | - Sara Succu
- Department of Veterinary Medicine, Anatomy, University of Sassari, 07100, Sassari, Italy
| | - Adele Frau
- Department of Veterinary Medicine, Obstetrics and Gynecology Clinics, University of Sassari, 07100, Sassari, Italy
| | - Francesca Mossa
- Department of Veterinary Medicine, Obstetrics and Gynecology Clinics, University of Sassari, 07100, Sassari, Italy
| | - Sylvia Virginie Versace
- Department of Veterinary Medicine, Veterinary Teaching Hospital, University of Sassari, 07100, Sassari, Italy
| | - Tiziana A L Brevini
- Laboratory of Biomedical Embryology, Department of Veterinary Medicine and Animal Science and Center for Stem Cell Research, University of Milano, 26900, Lodi, Italy
| | - Sergio Ledda
- Department of Veterinary Medicine, Obstetrics and Gynecology Clinics, University of Sassari, 07100, Sassari, Italy
| | - Daniela Bebbere
- Department of Veterinary Medicine, Obstetrics and Gynecology Clinics, University of Sassari, 07100, Sassari, Italy.
| |
Collapse
|
5
|
Stejskal S, Rájecká V, Covelo-Molares H, Sinigaglia K, Brožinová K, Kašiarová L, Dohnálková M, Reyes-Gutierrez PE, Cahová H, Keegan LP, O'Connell MA, Vaňáčová Š. Global analysis by LC-MS/MS of N6-methyladenosine and inosine in mRNA reveal complex incidence. RNA (NEW YORK, N.Y.) 2025; 31:514-528. [PMID: 39746750 PMCID: PMC11912911 DOI: 10.1261/rna.080324.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 12/05/2024] [Indexed: 01/04/2025]
Abstract
The precise and unambiguous detection and quantification of internal RNA modifications represents a critical step for understanding their physiological functions. The methods of direct RNA sequencing are quickly developing allowing for the precise location of internal RNA marks. This detection is, however, not quantitative and still presents detection limits. One of the biggest remaining challenges in the field is still the detection and quantification of m6A, m6Am, inosine, and m1A modifications of adenosine. The second intriguing and timely question remaining to be addressed is the extent to which individual marks are coregulated or potentially can affect each other. Here, we present a methodological approach to detect and quantify several key mRNA modifications in human total RNA and in mRNA, which is difficult to purify away from contaminating tRNA. We show that the adenosine demethylase FTO primarily targets m6Am marks in noncoding RNAs in HEK293T cells. Surprisingly, we observe little effect of FTO or ALKBH5 depletion on the m6A mRNA levels. Interestingly, the upregulation of ALKBH5 is accompanied by an increase in inosine level in overall mRNA.
Collapse
Affiliation(s)
- Stanislav Stejskal
- Central European Institute of Technology (CEITEC), Masaryk University, Brno 62500, Czech Republic
| | - Veronika Rájecká
- Central European Institute of Technology (CEITEC), Masaryk University, Brno 62500, Czech Republic
| | - Helena Covelo-Molares
- Central European Institute of Technology (CEITEC), Masaryk University, Brno 62500, Czech Republic
| | - Ketty Sinigaglia
- Central European Institute of Technology (CEITEC), Masaryk University, Brno 62500, Czech Republic
| | - Květoslava Brožinová
- Central European Institute of Technology (CEITEC), Masaryk University, Brno 62500, Czech Republic
| | - Linda Kašiarová
- Central European Institute of Technology (CEITEC), Masaryk University, Brno 62500, Czech Republic
| | - Michaela Dohnálková
- Central European Institute of Technology (CEITEC), Masaryk University, Brno 62500, Czech Republic
| | | | - Hana Cahová
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Liam P Keegan
- Central European Institute of Technology (CEITEC), Masaryk University, Brno 62500, Czech Republic
| | - Mary A O'Connell
- Central European Institute of Technology (CEITEC), Masaryk University, Brno 62500, Czech Republic
| | - Štěpánka Vaňáčová
- Central European Institute of Technology (CEITEC), Masaryk University, Brno 62500, Czech Republic
| |
Collapse
|
6
|
Chen W, Dong L, Wei C, Wu H. Role of epigenetic regulation in diminished ovarian reserve. J Assist Reprod Genet 2025; 42:389-403. [PMID: 39644448 PMCID: PMC11871224 DOI: 10.1007/s10815-024-03301-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 10/18/2024] [Indexed: 12/09/2024] Open
Abstract
Diminished ovarian reserve (DOR) is characterized by a decrease in the number and quality of oocytes, with its incidence increasing annually. Its pathogenesis remains unclear, making it one of the most challenging problems in the field of assisted reproduction. Epigenetic modification, a molecular mechanism affecting genomic activity and expression without altering the DNA sequence, has been widely studied in reproductive medicine and has attracted considerable attention regarding DOR. This review comprehensively examines the various epigenetic regulatory changes in ovarian granulosa cells (OGCs) and oocytes during DOR. DNA methylation plays a crucial role in regulating granulosa cell function, hormone production, and oocyte development, maturation, and senescence. Histone modifications are involved in regulating follicular activation, while non-coding RNAs, such as long noncoding RNAs (lncRNAs) and microRNAs (miRNAs), regulate granulosa cell function and oocyte development. N6-methyladenosine (m6A) modifications are associated with age-related oocyte senescence. Epigenetic clocks based on DNA methylation show potential in predicting ovarian reserve in DOR. Furthermore, it discusses the potential for utilizing epigenetic mechanisms to better diagnose and manage DOR.
Collapse
Affiliation(s)
- Wen Chen
- First School of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250011, China
| | - Li Dong
- First School of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250011, China
| | - Chaofeng Wei
- First School of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250011, China
| | - Haicui Wu
- Department of Reproduction and Genetics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China.
| |
Collapse
|
7
|
Fang M, Ye L, Zhu Y, Huang L, Xu S. M6A Demethylase ALKBH5 in Human Diseases: From Structure to Mechanisms. Biomolecules 2025; 15:157. [PMID: 40001461 PMCID: PMC11853652 DOI: 10.3390/biom15020157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/05/2025] [Accepted: 01/17/2025] [Indexed: 02/27/2025] Open
Abstract
N6-methyladenosine (m6A) is the most abundant, dynamically reversible, and evolutionarily conserved internal chemical modification in eukaryotic RNA. It is emerging as critical for regulating gene expression at the post-transcriptional level by affecting RNA metabolism through, for example, pre-mRNA processing, mRNA decay, and translation. ALKBH5 has recently been identified as an endogenous m6A demethylase implicated in a multitude of biological processes. This review provides an overview of the structural and functional characteristics of ALKBH5 and the involvement of ALKBH5 in diverse human diseases, including metabolic, immune, reproductive, and nervous system disorders, as well as the development of inhibitors. In summation, this review highlights the current understanding of the structure, functions, and detailed mechanisms of ALKBH5 in various physiological and pathological processes and provides valuable insights for clinical applications and foundational research within related fields.
Collapse
Affiliation(s)
| | | | | | | | - Shun Xu
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, Institute of Aging Research, School of Medical Technology, Guangdong Medical University, Songshan Lake, Dongguan 523808, China; (M.F.); (L.Y.); (Y.Z.); (L.H.)
| |
Collapse
|
8
|
Cui X, Li H, Huang X, Xue T, Wang S, Zhu X, Jing X. N 6-Methyladenosine Modification on the Function of Female Reproductive Development and Related Diseases. Immun Inflamm Dis 2024; 12:e70089. [PMID: 39660878 PMCID: PMC11632877 DOI: 10.1002/iid3.70089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 10/17/2024] [Accepted: 11/20/2024] [Indexed: 12/12/2024] Open
Abstract
BACKGROUND N6-methyladenosine (m6A) modification is a widespread and reversible epigenetic alteration in eukaryotic mRNA, playing a pivotal role in various biological functions. Its significance in female reproductive development and associated diseases has recently become a focal point of research. OBJECTIVE This review aims to consolidate current knowledge of the role of m6A modification in female reproductive tissues, emphasizing its regulatory dynamics, functional significance, and implications in reproductive health and disease. METHODS A comprehensive analysis of recent studies focusing on m6A modification in ovarian development, oocyte maturation, embryo development, and the pathogenesis of reproductive diseases. RESULTS m6A modification exhibits dynamic regulation in female reproductive tissues, influencing key developmental stages and processes. It plays critical roles in ovarian development, oocyte maturation, and embryo development, underpinning essential aspects of reproductive health. m6A modification is intricately involved in the pathogenesis of several reproductive diseases, including polycystic ovary syndrome (PCOS), premature ovarian failure (POF), and endometriosis, offering insights into potential molecular mechanisms and therapeutic targets. CONCLUSION The review highlights the crucial role of m6A modification in female reproductive development and related diseases. It underscores the need for further research to explore innovative diagnostic and therapeutic strategies for reproductive disorders, leveraging the insights gained from understanding m6A modification's impact on reproductive health.
Collapse
Affiliation(s)
- Xiangrong Cui
- Reproductive Medicine CenterThe affiliated Children's Hospital of Shanxi Medical University, Children's Hospital of Shanxi, Shanxi Maternal and Child Health HospitalTaiyuanChina
| | - Huihui Li
- Reproductive Medicine CenterThe affiliated Children's Hospital of Shanxi Medical University, Children's Hospital of Shanxi, Shanxi Maternal and Child Health HospitalTaiyuanChina
| | - Xia Huang
- Department of Clinical LaboratoryShanxi Provincial People's Hospital, Shanxi Medical UniversityTaiyuanChina
| | - Tingting Xue
- Department of Clinical LaboratoryShanxi Provincial People's Hospital, Shanxi Medical UniversityTaiyuanChina
| | - Shu Wang
- Department of Clinical LaboratoryShanxi Provincial People's Hospital, Shanxi Medical UniversityTaiyuanChina
| | - Xinyu Zhu
- Department of Clinical LaboratoryShanxi Provincial People's Hospital, Shanxi Medical UniversityTaiyuanChina
| | - Xuan Jing
- Department of Clinical LaboratoryShanxi Provincial People's Hospital, Shanxi Medical UniversityTaiyuanChina
| |
Collapse
|
9
|
Wen J, Zhu Q, Liu Y, Gou LT. RNA modifications: emerging players in the regulation of reproduction and development. Acta Biochim Biophys Sin (Shanghai) 2024; 57:33-58. [PMID: 39574165 PMCID: PMC11802351 DOI: 10.3724/abbs.2024201] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 11/05/2024] [Indexed: 01/25/2025] Open
Abstract
The intricate world of RNA modifications, collectively termed the epitranscriptome, covers over 170 identified modifications and impacts RNA metabolism and, consequently, almost all biological processes. In this review, we focus on the regulatory roles and biological functions of a panel of dominant RNA modifications (including m 6A, m 5C, Ψ, ac 4C, m 1A, and m 7G) on three RNA types-mRNA, tRNA, and rRNA-in mammalian development, particularly in the context of reproduction as well as embryonic development. We discuss in detail how those modifications, along with their regulatory proteins, affect RNA processing, structure, localization, stability, and translation efficiency. We also highlight the associations among dysfunctions in RNA modification-related proteins, abnormal modification deposition and various diseases, emphasizing the roles of RNA modifications in critical developmental processes such as stem cell self-renewal and cell fate transition. Elucidating the molecular mechanisms by which RNA modifications influence diverse developmental processes holds promise for developing innovative strategies to manage developmental disorders. Finally, we outline several unexplored areas in the field of RNA modification that warrant further investigation.
Collapse
Affiliation(s)
- Junfei Wen
- Key Laboratory of RNA InnovationScience and EngineeringShanghai Key Laboratory of Molecular AndrologyCAS Center for Excellence in Molecular. Cell ScienceShanghai Institute of Biochemistry and Cell BiologyChinese Academy of SciencesShanghai200031China
- University of Chinese Academy of SciencesBeijing100049China
| | - Qifan Zhu
- Key Laboratory of RNA InnovationScience and EngineeringShanghai Key Laboratory of Molecular AndrologyCAS Center for Excellence in Molecular. Cell ScienceShanghai Institute of Biochemistry and Cell BiologyChinese Academy of SciencesShanghai200031China
- University of Chinese Academy of SciencesBeijing100049China
| | - Yong Liu
- Key Laboratory of RNA InnovationScience and EngineeringShanghai Key Laboratory of Molecular AndrologyCAS Center for Excellence in Molecular. Cell ScienceShanghai Institute of Biochemistry and Cell BiologyChinese Academy of SciencesShanghai200031China
| | - Lan-Tao Gou
- Key Laboratory of RNA InnovationScience and EngineeringShanghai Key Laboratory of Molecular AndrologyCAS Center for Excellence in Molecular. Cell ScienceShanghai Institute of Biochemistry and Cell BiologyChinese Academy of SciencesShanghai200031China
- University of Chinese Academy of SciencesBeijing100049China
| |
Collapse
|
10
|
Li D, Hu S, Ye J, Zhai C, Liu J, Wang Z, Zhou X, Chen L, Zhou F. The Emerging Role of IGF2BP2 in Cancer Therapy Resistance: From Molecular Mechanism to Future Potential. Int J Mol Sci 2024; 25:12150. [PMID: 39596216 PMCID: PMC11595103 DOI: 10.3390/ijms252212150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 11/04/2024] [Accepted: 11/06/2024] [Indexed: 11/28/2024] Open
Abstract
Tumor resistance is one of the primary reasons for cancer treatment failure, significantly limiting the options and efficacy of cancer therapies. Therefore, overcoming resistance has become a critical factor in improving cancer treatment outcomes. IGF2BP2, as a reader of m6A methylation, plays a pivotal role in the post-transcriptional regulation of RNA through the methylation of m6A sites. It not only contributes to cancer initiation and progression but also plays a key role in tumor drug resistance. This review provides a comprehensive summary of the mechanisms by which IGF2BP2 contributes to therapy resistance, with the aim of improving the efficacy of chemotherapy in cancer treatment. Advancing research in this area is crucial for developing more effective therapies that could significantly improve the quality of life for cancer patients.
Collapse
Affiliation(s)
- Die Li
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China; (D.L.); (S.H.); (J.Y.); (C.Z.); (J.L.); (Z.W.); (X.Z.)
| | - Shiqi Hu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China; (D.L.); (S.H.); (J.Y.); (C.Z.); (J.L.); (Z.W.); (X.Z.)
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200433, China
| | - Jiarong Ye
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China; (D.L.); (S.H.); (J.Y.); (C.Z.); (J.L.); (Z.W.); (X.Z.)
| | - Chaojie Zhai
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China; (D.L.); (S.H.); (J.Y.); (C.Z.); (J.L.); (Z.W.); (X.Z.)
| | - Jipeng Liu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China; (D.L.); (S.H.); (J.Y.); (C.Z.); (J.L.); (Z.W.); (X.Z.)
| | - Zuao Wang
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China; (D.L.); (S.H.); (J.Y.); (C.Z.); (J.L.); (Z.W.); (X.Z.)
| | - Xinchi Zhou
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China; (D.L.); (S.H.); (J.Y.); (C.Z.); (J.L.); (Z.W.); (X.Z.)
| | - Leifeng Chen
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
- Medical Center for Cardiovascular Diseases, Neurological Diseases and Tumors of Jiangxi Province, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Fan Zhou
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China; (D.L.); (S.H.); (J.Y.); (C.Z.); (J.L.); (Z.W.); (X.Z.)
| |
Collapse
|
11
|
Zhou Q, Liu X, Lu H, Li N, Meng J, Huang J, Zhang Z, Liu J, Fan W, Li W, Li X, Liu X, Zuo H, Yang P, Hou S. m6A-methylase METTL3 promotes retinal angiogenesis through modulation of metabolic reprogramming in RPE cells. J Neuroinflammation 2024; 21:289. [PMID: 39506758 PMCID: PMC11539582 DOI: 10.1186/s12974-024-03279-1] [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: 09/13/2024] [Accepted: 10/28/2024] [Indexed: 11/08/2024] Open
Abstract
Retinal neovascularization (RNV) disease is one of the leading causes of blindness, yet the molecular underpinnings of this condition are not well understood. To delve into the critical aspects of cell-mediated angiogenesis, we analyzed our previously published single-cell data. Our analysis revealed that retinal pigment epithelium (RPE) cells serve a crucial promotional function in angiogenesis. RPE cells were regulated by N6-methyladenosine (m6A). Next, we detected several critical m6A methylase in hypoxic ARPE-19 cells and in oxygen-induced retinopathy (OIR) mice, our results revealed a significant decrease in the level of methyltransferase like 3 (METTL3). METTL3 specific inhibitor STM2457 intravitreal injection or METTL3 conditional knockout mice both showed a significantly reduced neovascularization area of retina. Additionally, the angiogenesis-related abilities of human retinal endothelial cells (HRECs) were diminished after co-cultured with ARPE-19 treated with STM2457 or sh-METTL3 in vitro. Furthermore, through the integration of Methylated RNA immunoprecipitation (MeRIP) sequencing and RNA sequencing, we discovered that the metabolic enzyme quinolinate phosphoribosyltransferase (QPRT) was directly modified by METTL3 and recognized by the YTH N6-methyladenosine RNA binding protein C1 (YTHDC1). Moreover, after over-expressing QPRT, the angiogenic abilities of HRECs were improved through the phosphorylated phosphatidylinositol-3-kinase (p-PI3K)/ phosphorylated threonine kinase (p-AKT) pathway. Collectively, our study provided a novel therapeutic target for retinal angiogenesis.
Collapse
Affiliation(s)
- Qian Zhou
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, and Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing, 400016, China
| | - Xianyang Liu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, and Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing, 400016, China
- Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Huiping Lu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, and Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing, 400016, China
| | - Na Li
- Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Jiayu Meng
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Jiaxing Huang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, and Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing, 400016, China
| | - Zhi Zhang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, and Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing, 400016, China
| | - Jiangyi Liu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, and Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing, 400016, China
| | - Wei Fan
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, and Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing, 400016, China
| | - Wanqian Li
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, and Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing, 400016, China
| | - Xingran Li
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, and Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing, 400016, China
| | - Xiaoyan Liu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, and Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing, 400016, China
| | - Hangjia Zuo
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, and Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing, 400016, China
| | - Peizeng Yang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, and Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing, 400016, China.
| | - Shengping Hou
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, and Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing, 400016, China.
- Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China.
| |
Collapse
|
12
|
Shi JX, Zhang ZC, Yin HZ, Piao XJ, Liu CH, Liu QJ, Zhang JC, Zhou WX, Liu FC, Yang F, Wang YF, Liu H. RNA m6A modification in ferroptosis: implications for advancing tumor immunotherapy. Mol Cancer 2024; 23:213. [PMID: 39342168 PMCID: PMC11437708 DOI: 10.1186/s12943-024-02132-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 09/19/2024] [Indexed: 10/01/2024] Open
Abstract
The pursuit of innovative therapeutic strategies in oncology remains imperative, given the persistent global impact of cancer as a leading cause of mortality. Immunotherapy is regarded as one of the most promising techniques for systemic cancer therapies among the several therapeutic options available. Nevertheless, limited immune response rates and immune resistance urge us on an augmentation for therapeutic efficacy rather than sticking to conventional approaches. Ferroptosis, a novel reprogrammed cell death, is tightly correlated with the tumor immune environment and interferes with cancer progression. Highly mutant or metastasis-prone tumor cells are more susceptible to iron-dependent nonapoptotic cell death. Consequently, ferroptosis-induction therapies hold the promise of overcoming resistance to conventional treatments. The most prevalent post-transcriptional modification, RNA m6A modification, regulates the metabolic processes of targeted RNAs and is involved in numerous physiological and pathological processes. Aberrant m6A modification influences cell susceptibility to ferroptosis, as well as the expression of immune checkpoints. Clarifying the regulation of m6A modification on ferroptosis and its significance in tumor cell response will provide a distinct method for finding potential targets to enhance the effectiveness of immunotherapy. In this review, we comprehensively summarized regulatory characteristics of RNA m6A modification on ferroptosis and discussed the role of RNA m6A-mediated ferroptosis on immunotherapy, aiming to enhance the effectiveness of ferroptosis-sensitive immunotherapy as a treatment for immune-resistant malignancies.
Collapse
Affiliation(s)
- Jun-Xiao Shi
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Zhi-Chao Zhang
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Hao-Zan Yin
- The Department of Medical Genetics, Naval Medical University, Shanghai, 200433, China
| | - Xian-Jie Piao
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Cheng-Hu Liu
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Qian-Jia Liu
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Jia-Cheng Zhang
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Wen-Xuan Zhou
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Fu-Chen Liu
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Fu Yang
- The Department of Medical Genetics, Naval Medical University, Shanghai, 200433, China.
- Key Laboratory of Biosafety Defense, Ministry of Education, Shanghai, 200433, China.
- Shanghai Key Laboratory of Medical Biodefense, Shanghai, 200433, China.
| | - Yue-Fan Wang
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China.
| | - Hui Liu
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China.
| |
Collapse
|
13
|
Gao Z, Zha X, Li M, Xia X, Wang S. Insights into the m 6A demethylases FTO and ALKBH5 : structural, biological function, and inhibitor development. Cell Biosci 2024; 14:108. [PMID: 39192357 DOI: 10.1186/s13578-024-01286-6] [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: 05/22/2024] [Accepted: 08/13/2024] [Indexed: 08/29/2024] Open
Abstract
N6-methyladenosine (m6A) is dynamically regulated by methyltransferases (termed "writers") and demethylases (referred to as "erasers"), facilitating a reversible modulation. Changes in m6A levels significantly influence cellular functions, such as RNA export from the nucleus, mRNA metabolism, protein synthesis, and RNA splicing. They are intricately associated with a spectrum of pathologies. Moreover, dysregulation of m6A modulation has emerged as a promising therapeutic target across many diseases. m6A plays a pivotal role in controlling vital downstream molecules and critical biological pathways, contributing to the pathogenesis and evolution of numerous conditions. This review provides an overview of m6A demethylases, explicitly detailing the structural and functional characteristics of FTO and ALKBH5. Additionally, we explore their distinct involvement in various diseases, examine factors regulating their expression, and discuss the progress in inhibitor development.
Collapse
Affiliation(s)
- Zewei Gao
- Department of Laboratory Medicine,Jiangsu Province Engineering Research Center for Precise Diagnosis and Treatment of Inflammatory Diseases, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
- Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Xuan Zha
- Department of Laboratory Medicine,Jiangsu Province Engineering Research Center for Precise Diagnosis and Treatment of Inflammatory Diseases, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
- Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Min Li
- Department of Laboratory Medicine, Affiliated People's Hospital, Jiangsu University, Zhenjiang, 212002, China.
| | - Xueli Xia
- Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Shengjun Wang
- Department of Laboratory Medicine,Jiangsu Province Engineering Research Center for Precise Diagnosis and Treatment of Inflammatory Diseases, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China.
- Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China.
| |
Collapse
|
14
|
Wells GR, Pillai RS. Roles of N 6-methyladenosine writers, readers and erasers in the mammalian germline. Curr Opin Genet Dev 2024; 87:102224. [PMID: 38981182 DOI: 10.1016/j.gde.2024.102224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 06/06/2024] [Accepted: 06/23/2024] [Indexed: 07/11/2024]
Abstract
N6-methyladenosine (m6A) is the most abundant internal modification of mRNAs in eukaryotes. Numerous studies have shown that m6A plays key roles in many biological and pathophysiological processes, including fertility. The factors involved in m6A-dependent mRNA regulation include writers, which deposit the m6A mark, erasers, which remove it, and readers, which bind to m6A-modified transcripts and mediate the regulation of mRNA fate. Many of these proteins are highly expressed in the germ cells of mammals, and some have been linked to fertility disorders in human patients. In this review, we summarise recent findings on the important roles played by proteins involved in m6A biology in mammalian gametogenesis and fertility. Continued study of the m6A pathway in the mammalian germline will shed further light on the importance of epitranscriptomics in reproduction and may lead to effective treatment of human fertility disorders.
Collapse
Affiliation(s)
- Graeme R Wells
- Department of Molecular Biology, Science III, University of Geneva, 30 Quai Ernest-Ansermet, CH-1211 Geneva 4, Switzerland.
| | - Ramesh S Pillai
- Department of Molecular Biology, Science III, University of Geneva, 30 Quai Ernest-Ansermet, CH-1211 Geneva 4, Switzerland
| |
Collapse
|
15
|
Wei G. RNA m6A modification, signals for degradation or stabilisation? Biochem Soc Trans 2024; 52:707-717. [PMID: 38629637 PMCID: PMC11088905 DOI: 10.1042/bst20230574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 03/24/2024] [Accepted: 04/08/2024] [Indexed: 04/25/2024]
Abstract
The RNA modification N6-methyladenosine (m6A) is conserved across eukaryotes, and profoundly influences RNA metabolism, including regulating RNA stability. METTL3 and METTL14, together with several accessory components, form a 'writer' complex catalysing m6A modification. Conversely, FTO and ALKBH5 function as demethylases, rendering m6A dynamic. Key to understanding the functional significance of m6A is its 'reader' proteins, exemplified by YTH-domain-containing proteins (YTHDFs) canonical reader and insulin-like growth factor 2 mRNA-binding proteins (IGF2BPs) non-canonical reader. These proteins play a crucial role in determining RNA stability: YTHDFs mainly promote mRNA degradation through different cytoplasmic pathways, whereas IGF2BPs function to maintain mRNA stability. Additionally, YTHDC1 functions within the nucleus to degrade or protect certain m6A-containing RNAs, and other non-canonical readers also contribute to RNA stability regulation. Notably, m6A regulates retrotransposon LINE1 RNA stability and/or transcription via multiple mechanisms. However, conflicting observations underscore the complexities underlying m6A's regulation of RNA stability depending upon the RNA sequence/structure context, developmental stage, and/or cellular environment. Understanding the interplay between m6A and other RNA regulatory elements is pivotal in deciphering the multifaceted roles m6A plays in RNA stability regulation and broader cellular biology.
Collapse
Affiliation(s)
- Guifeng Wei
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, U.K
| |
Collapse
|