1
|
Ma X, Li H, Zhao Z, Li C, Wang M, Zhang L, Zhao Y, Su H, Wang F, Hua J. The HNRNPC/CELF2 signaling pathway drives glycolytic reprogramming and mitochondrial dysfunction in drug-resistant acute myeloid leukemia. Cell Biosci 2025; 15:61. [PMID: 40380235 PMCID: PMC12083169 DOI: 10.1186/s13578-025-01386-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 03/29/2025] [Indexed: 05/19/2025] Open
Abstract
BACKGROUND Acute myeloid leukemia (AML) is an aggressive cancer with high treatment resistance, often leading to poor patient outcomes. Metabolic reprogramming plays a critical role in AML progression, influencing drug resistance (DR) and tumor survival. This study investigates the HNRNPC/CELF2 signaling pathway and its impact on AML cell metabolism and DR. RESULTS The study identified that HNRNPC regulates the expression of CELF2 through m6 A modification. In drug-resistant AML cells, increased HNRNPC expression and decreased CELF2 expression were associated with upregulated glycolysis, enhanced glucose consumption, lactate production, and mitochondrial dysfunction. Knockdown of HNRNPC reduced glycolysis and cell invasion, while CELF2 knockdown reversed these effects. Conversely, HNRNPC overexpression enhanced glycolysis and cell migration, which were counteracted by CELF2 overexpression. CONCLUSIONS The HNRNPC/CELF2 axis plays a pivotal role in metabolic reprogramming, driving AML progression and chemotherapy resistance. Targeting this pathway may offer new therapeutic strategies to overcome resistance and improve treatment outcomes in AML patients.
Collapse
Affiliation(s)
- Xiang Ma
- Laboratory of Biochemistry and Pharmacy, Taiyuan Institute of Technology, No. 31, Xinlan Road, Jiancaoping District, Taiyuan, 030008, Shanxi, People's Republic of China
| | - Haodong Li
- Laboratory of Biochemistry and Pharmacy, Taiyuan Institute of Technology, No. 31, Xinlan Road, Jiancaoping District, Taiyuan, 030008, Shanxi, People's Republic of China
| | - Ziqi Zhao
- Chemistry and Chemical Engineering Department, Taiyuan Institute of Technology, Taiyuan, 030008, People's Republic of China
| | - Changchun Li
- Chemistry and Chemical Engineering Department, Taiyuan Institute of Technology, Taiyuan, 030008, People's Republic of China
| | - Man Wang
- Laboratory of Biochemistry and Pharmacy, Taiyuan Institute of Technology, No. 31, Xinlan Road, Jiancaoping District, Taiyuan, 030008, Shanxi, People's Republic of China
| | - Lele Zhang
- Laboratory of Biochemistry and Pharmacy, Taiyuan Institute of Technology, No. 31, Xinlan Road, Jiancaoping District, Taiyuan, 030008, Shanxi, People's Republic of China
| | - Yutong Zhao
- Laboratory of Biochemistry and Pharmacy, Taiyuan Institute of Technology, No. 31, Xinlan Road, Jiancaoping District, Taiyuan, 030008, Shanxi, People's Republic of China
| | - Haipeng Su
- Laboratory of Biochemistry and Pharmacy, Taiyuan Institute of Technology, No. 31, Xinlan Road, Jiancaoping District, Taiyuan, 030008, Shanxi, People's Republic of China
| | - Feng Wang
- Chemistry and Chemical Engineering Department, Taiyuan Institute of Technology, Taiyuan, 030008, People's Republic of China
| | - Jiai Hua
- Laboratory of Biochemistry and Pharmacy, Taiyuan Institute of Technology, No. 31, Xinlan Road, Jiancaoping District, Taiyuan, 030008, Shanxi, People's Republic of China.
- Chemistry and Chemical Engineering Department, Taiyuan Institute of Technology, Taiyuan, 030008, People's Republic of China.
| |
Collapse
|
2
|
Leng Y, Luan Z, Li Z, Ma Y, Zhou Y, Liu J, Liu S, Tian T, Feng W, Liu Y, Shi Q, Huang C, Zhao X, Wang W, Liu A, Wang T, Ren Q, Liu J, Huang Q, Zhang Y, Yin B, Chen J, Yang L, Zhao S, Bao R, Ji X, Xu Y, Liu L, Zhou J, Chen M, Ma W, Shen L, Zhang T, Zhao H. PPM1F regulates ovarian cancer progression by affecting the dephosphorylation of ITGB1. Clin Transl Oncol 2025; 27:1013-1025. [PMID: 39133386 DOI: 10.1007/s12094-024-03614-1] [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: 04/29/2024] [Accepted: 07/09/2024] [Indexed: 08/13/2024]
Abstract
PPM1F has been shown to play diverse biological functions in the progression of multiple tumors. PPM1F controls the T788/T789 phosphorylation switch of ITGB1 and regulates integrin activity. However, the impacts of PPM1F and ITGB1 on ovarian cancer (OV) progression remain unclear. Whether there is such a regulatory relationship between PPM1F and ITGB1 in ovarian cancer has not been studied. Therefore, the purpose of this study is to elucidate the function and the mechanism of PPM1F in ovarian cancer. The expression level and the survival curve of PPM1F were analyzed by databases. Gain of function and loss of function were applied to explore the function of PPM1F in ovarian cancer. A tumor formation assay in nude mice showed that knockdown of PPM1F inhibited tumor formation. We tested the effect of PPM1F on ITGB1 dephosphorylation in ovarian cancer cells by co-immunoprecipitation and western blotting. Loss of function was applied to investigate the function of ITGB1 in ovarian cancer. ITGB1-mut overexpression promotes the progression of ovarian cancer. Rescue assays showed the promoting effect of ITGB1-wt on ovarian cancer is attenuated due to the dephosphorylation of ITGB1-wt by PPM1F. PPM1F and ITGB1 play an oncogene function in ovarian cancer. PPM1F regulates the phosphorylation of ITGB1, which affects the occurrence and development of ovarian cancer.
Collapse
Affiliation(s)
- Yahui Leng
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China
| | - Zhenzi Luan
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China
| | - Zihang Li
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China
| | - Yongqing Ma
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China
| | - Yang Zhou
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China
| | - Jiaqi Liu
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China
| | - Song Liu
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China
| | - Tian Tian
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China
| | - Wenxiao Feng
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China
| | - Yanni Liu
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China
| | - Qin Shi
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China
| | - Chengyang Huang
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China
| | - Xuan Zhao
- The Second Clinical College, Xi'an Medical University, Xi'an, China
| | - Wenlong Wang
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China
| | - Ao Liu
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China
| | - Tianhang Wang
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China
| | - Qiulei Ren
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China
| | - Jiakun Liu
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China
| | - Qian Huang
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China
| | - Yaling Zhang
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China
| | - Bin Yin
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China
| | - Jialin Chen
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China
| | - Liangliang Yang
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China
| | - Shiyun Zhao
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China
| | - Ruoyi Bao
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China
| | - Xingyu Ji
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China
| | - Yuewen Xu
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China
| | - Liaoyuan Liu
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China
| | - Junsuo Zhou
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China
| | - Miao Chen
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China
| | - Wenhui Ma
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China
| | - Li Shen
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China.
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China.
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China.
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China.
| | - Te Zhang
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China.
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China.
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China.
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China.
| | - Hongyan Zhao
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China.
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China.
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China.
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China.
| |
Collapse
|
3
|
Tang J, Zhou C, Ye F, Zuo S, Zhou M, Lu L, Chai P, Fan X. RNA methylation homeostasis in ocular diseases: All eyes on Me. Prog Retin Eye Res 2025; 105:101335. [PMID: 39880118 DOI: 10.1016/j.preteyeres.2025.101335] [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: 07/28/2024] [Revised: 01/22/2025] [Accepted: 01/23/2025] [Indexed: 01/31/2025]
Abstract
RNA methylation is a pivotal epigenetic modification that adjusts various aspects of RNA biology, including nuclear transport, stability, and the efficiency of translation for specific RNA candidates. The methylation of RNA involves the addition of methyl groups to specific bases and can occur at different sites, resulting in distinct forms, such as N6-methyladenosine (m6A), N1-methyladenosine (m1A), 5-methylcytosine (m5C), and 7-methylguanosine (m7G). Maintaining an optimal equilibrium of RNA methylation is crucial for fundamental cellular activities such as cell survival, proliferation, and migration. The balance of RNA methylation is linked to various pathophysiological conditions, including senescence, cancer development, stress responses, and blood vessel formation, all of which are pivotal for comprehending a spectrum of eye diseases. Recent findings have highlighted the significant role of diverse RNA methylation patterns in ophthalmological conditions such as age-related macular degeneration, diabetic retinopathy, cataracts, glaucoma, uveitis, retinoblastoma, uveal melanoma, thyroid eye disease, and myopia, which are critical for vision health. This thorough review endeavors to dissect the influence of RNA methylation on common and vision-impairing ocular disorders. It explores the nuanced roles that RNA methylation plays in key pathophysiological mechanisms, such as oxidative stress and angiogenesis, which are integral to the onset and progression of these diseases. By synthesizing the latest research, this review offers valuable insights into how RNA methylation could be harnessed for therapeutic interventions in the field of ophthalmology.
Collapse
Affiliation(s)
- Jieling Tang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, State Key Laboratory of Vision Health, China
| | - Chuandi Zhou
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, State Key Laboratory of Vision Health, China
| | - Fuxiang Ye
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, State Key Laboratory of Vision Health, China
| | - Sipeng Zuo
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, State Key Laboratory of Vision Health, China
| | - Min Zhou
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, State Key Laboratory of Vision Health, China
| | - Linna Lu
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, State Key Laboratory of Vision Health, China.
| | - Peiwei Chai
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, State Key Laboratory of Vision Health, China.
| | - Xianqun Fan
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, State Key Laboratory of Vision Health, China.
| |
Collapse
|
4
|
Han N, Yu N, Yu L. The mRNA Stability of PIEZO1, Regulated by Methyltransferase-Like 3 via N 6-Methylation of Adenosine Modification in a YT521-B Homology Domain Family 2-Dependent Manner, Facilitates the Progression of Diabetic Retinopathy. THE AMERICAN JOURNAL OF PATHOLOGY 2025; 195:265-280. [PMID: 39476953 DOI: 10.1016/j.ajpath.2024.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 09/29/2024] [Accepted: 10/10/2024] [Indexed: 11/09/2024]
Abstract
Diabetic retinopathy (DR) is the major ocular complication of diabetes caused by chronic hyperglycemia, which leads to incurable blindness. Currently, the effectiveness of therapeutic interventions is limited. This study aimed to investigate the function of piezo-type mechanosensitive ion channel component 1 (PIEZO1) and its potential regulatory mechanism in DR progression. PIEZO1 expression was up-regulated in the retinal tissues of streptozotocin-induced diabetic mice and high-glucose (HG)-triggered Müller cells. Functionally, the knockdown of PIEZO1 improved the abnormal retinal function of diabetic mice and impeded inflammatory cytokine secretion and gliosis of Müller cells under HG conditions. Mechanistic investigations using RNA immunoprecipitation-real-time quantitative PCR, methylation RNA immunoprecipitation-real-time quantitative PCR, and luciferase reporter assays demonstrated that PIEZO1 was a downstream target of methyltransferase-like 3 (METTL3). METTL3-mediated N6-methyladenosine (m6A) modification within the coding sequence of PIEZO1 mRNA significantly shortened its half-life. In HG-stimulated cells, there was a negative regulatory relationship between PIEZO1 and YTH (YT521-B homology) domain family 2 (YTHDF2), a recognized m6A reader. The loss of YTHDF2 resulted in an extended half-life of PIEZO1 in cells with overexpression of METTL3, indicating that the effect of METTL3 on the mRNA stability of PIEZO1 was dependent on YTHDF2. Taken together, this study demonstrated the protective role of the PIEZO1 silencing in DR development, and that the degradation of PIEZO1 mRNA is accelerated by METTL3/YTHDF2-mediated m6A modification.
Collapse
Affiliation(s)
- Ning Han
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, China
| | - Na Yu
- Department of Blood Transfusion, The Second Hospital of Jilin University, Changchun, China
| | - Li Yu
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, China.
| |
Collapse
|
5
|
Feng S, Zhang Q, Liu Q, Huang C, Zhang H, Wang F, Zhu Y, Jian Q, Chen X, Jiang Q, Yan B. N 6-Methyladenosine Demethylase FTO Controls Macrophage Homeostasis in Diabetic Vasculopathy. Diabetes 2025; 74:82-95. [PMID: 39446524 DOI: 10.2337/db24-0691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Accepted: 10/18/2024] [Indexed: 10/26/2024]
Abstract
Diabetic vasculopathy, encompassing complications such as diabetic retinopathy, represents a significant source of morbidity, with inflammation playing a pivotal role in the progression of these complications. This study investigates the influence of N6-methyladenosine demethylase (m6A) modification and the m6A demethylase fat mass and obesity-associated (FTO) protein on macrophage polarization and its subsequent effects on diabetic microvasculopathy. We found that diabetes induces a shift in macrophage polarization toward a proinflammatory M1 phenotype, which is associated with a reduction in m6A modification levels. Notably, FTO emerges as a critical regulator of m6A under diabetic conditions. In vitro experiments reveal that FTO not only modulates macrophage polarization but also mediates their interactions with vascular endothelial cells. In vivo experiments demonstrate that FTO deficiency exacerbates retinal inflammation and microvascular dysfunction in diabetic retinas. Mechanistically, FTO stabilizes mRNA through an m6A-YTHDF2-dependent pathway, thereby activating the PI3K/AKT signaling cascade. Collectively, these findings position FTO as a promising therapeutic target for the management of diabetic vascular complications. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Siguo Feng
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Qiuyang Zhang
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Qing Liu
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Chang Huang
- Eye Institute and Department of Ophthalmology, Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Huiying Zhang
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Fengsheng Wang
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Yue Zhu
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Qizhi Jian
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xue Chen
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Qin Jiang
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Biao Yan
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
6
|
Wu J, Chen Y, Shi S, Liu J, Zhang F, Li X, Liu X, Hu G, Dong Y. Exploration of Pharmacological Mechanisms of Dapagliflozin against Type 2 Diabetes Mellitus through PI3K-Akt Signaling Pathway based on Network Pharmacology Analysis and Deep Learning Technology. Curr Comput Aided Drug Des 2025; 21:452-465. [PMID: 38204223 DOI: 10.2174/0115734099274407231207070451] [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: 08/14/2023] [Revised: 10/20/2023] [Accepted: 11/01/2023] [Indexed: 01/12/2024]
Abstract
BACKGROUND Dapagliflozin is commonly used to treat type 2 diabetes mellitus (T2DM). However, research into the specific anti-T2DM mechanisms of dapagliflozin remains scarce. OBJECTIVE This study aimed to explore the underlying mechanisms of dapagliflozin against T2DM. METHODS Dapagliflozin-associated targets were acquired from CTD, SwissTargetPrediction, and SuperPred. T2DM-associated targets were obtained from GeneCards and DigSee. VennDiagram was used to obtain the overlapping targets of dapagliflozin and T2DM. GO and KEGG analyses were performed using clusterProfiler. A PPI network was built by STRING database and Cytoscape, and the top 30 targets were screened using the degree, maximal clique centrality (MCC), and edge percolated component (EPC) algorithms of CytoHubba. The top 30 targets screened by the three algorithms were intersected with the core pathway-related targets to obtain the key targets. DeepPurpose was used to evaluate the binding affinity of dapagliflozin with the key targets. RESULTS In total, 155 overlapping targets of dapagliflozin and T2DM were obtained. GO and KEGG analyses revealed that the targets were primarily enriched in response to peptide, membrane microdomain, protein serine/threonine/tyrosine kinase activity, PI3K-Akt signaling pathway, MAPK signaling pathway, and AGE-RAGE signaling pathway in diabetic complications. AKT1, PIK3CA, NOS3, EGFR, MAPK1, MAPK3, HSP90AA1, MTOR, RELA, NFKB1, IKBKB, ITGB1, and TP53 were the key targets, mainly related to oxidative stress, endothelial function, and autophagy. Through the DeepPurpose algorithm, AKT1, HSP90AA1, RELA, ITGB1, and TP53 were identified as the top 5 anti-targets of dapagliflozin. CONCLUSION Dapagliflozin might treat T2DM mainly by targeting AKT1, HSP90AA1, RELA, ITGB1, and TP53 through PI3K-Akt signaling.
Collapse
Affiliation(s)
- Jie Wu
- Department of Cardiology, Jinhua People's Hospital, Jinhua, Zhejiang, China
| | - Yufan Chen
- Department of Blood Donation Service, Central Blood Station of Jinhua, Jinhua, Zhejiang, China
| | - Shuai Shi
- Department of IVF, Jinhua People's Hospital, Jinhua, Zhejiang, China
| | - Junru Liu
- Department of Endocrinology, Jinhua People's Hospital, Jinhua, Zhejiang, China
| | - Fen Zhang
- Department of Cardiology, Jinhua People's Hospital, Jinhua, Zhejiang, China
| | - Xingxing Li
- Department of Cardiology, Jinhua People's Hospital, Jinhua, Zhejiang, China
| | - Xizhi Liu
- Department of Cardiology, Jinhua People's Hospital, Jinhua, Zhejiang, China
| | - Guoliang Hu
- Department of Ultrasound in Medicine, Jinhua People's Hospital, Jinhua, Zhejiang, China
| | - Yang Dong
- Department of Cardiology, Jinhua People's Hospital, Jinhua, Zhejiang, China
| |
Collapse
|
7
|
Liu S, Cao Y, Zhang Y. Regulatory roles of RNA methylation in vascular lesions in ocular and cardiopulmonary diseases. Crit Rev Clin Lab Sci 2024; 61:726-740. [PMID: 38957015 DOI: 10.1080/10408363.2024.2370267] [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: 12/28/2023] [Revised: 05/12/2024] [Accepted: 06/17/2024] [Indexed: 07/04/2024]
Abstract
RNA methylation is a widespread regulatory mechanism that controls gene expression in physiological processes. In recent years, the mechanisms and functions of RNA methylation under diseased conditions have been increasingly unveiled by RNA sequencing technologies with large scale and high resolution. In this review, the fundamental concept of RNA methylation is introduced, and the common types of transcript methylation and their machineries are described. Then, the regulatory roles of RNA methylation, particularly N6-methyladenosine and 5-methylcytosine, in the vascular lesions of ocular and cardiopulmonary diseases are discussed and compared. The ocular diseases include corneal neovascularization, retinopathy of prematurity, diabetic retinopathy, and pathologic myopia; whereas the cardiopulmonary ailments involve atherosclerosis and pulmonary hypertension. This review hopes to shed light on the common regulatory mechanisms underlying the vascular lesions in these ocular and cardiopulmonary diseases, which may be conducive to developing therapeutic strategies in clinical practice.
Collapse
Affiliation(s)
- Siyi Liu
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China
| | - Yunshan Cao
- Department of Cardiology, Gansu Provincial Hospital, Lanzhou, China
| | - Yan Zhang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China
| |
Collapse
|
8
|
Gong Q, Hu L, Liu G, Yin X, Zhao X, Li Q, Li Y, Sun Y, Zhou Y, Guo C, Du Z. WTAP-mediated N6-methyladenosine mRNA methylation regulates laser-induced macular neovascularization. Mol Vis 2024; 30:336-347. [PMID: 40330498 PMCID: PMC12055036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 10/05/2024] [Indexed: 05/08/2025] Open
Abstract
Purpose Neovascular age-related macular degeneration (nAMD) is now a major cause of central vision loss in older adults worldwide. The primary characteristic of nAMD is the formation of macular neovascularization (MNV), which is a pathologic form of angiogenesis. Epigenetics plays a role in multiple pathological physiologic processes. N6-methyladenosine (m6A) modification is the most common, abundant, and reversible modification in eukaryotic mRNAs, and it plays a role in various pathological angiogenesis processes. This study intends to reveal the expression and functions of m6A during the macular neovascularization (MNV) process. Methods A laser-induced MNV mouse model was used in this study. m6A quantitative analysis was performed to detect the expression of m6A. Subsequently, the expression of various m6A writers and erasers was detected using quantitative real-time polymerase chain reaction (qRT-PCR) and western blot. Immunohistochemistry was used to detect Wilms' tumor 1-associating protein (WTAP) expression in the MNV lesions. Intravitreal injection of WTAP siRNA in MNV mice to silence the WTAP gene. Hematoxylin and eosin (H&E) were used to determine the thickness and length of the MNV. Fundus fluorescein angiography (FFA) and indocyanine green angiography (ICGA) were examined to measure the leakage area of the MNV. Proliferating cell nuclear antigen (PCNA) expression was detected with a western blot. The mRNA and protein levels of β-catenin were tested with qRT-PCR and western blot. Results We found increased m6A modification levels after laser induction compared with the normal control group. Subsequently, the expression of various m6A writers and erasers was detected. The results showed that WTAP increased in the MNV model in mice. After the injection of WTAP siRNA into the vitreous body, the expression of WTAP significantly decreased, subsequently decreasing the m6A modification levels. The width, breadth, and leakage area of MNV damage markedly decreased, and endothelial cell proliferation was inhibited. After laser-induced MNV, the expression of β-catenin increased, and that of β-catenin significantly decreased after WTAP knockout. Conclusions In conclusion, this study suggests that WTAP-mediated m6A methylation can regulate pathological angiogenesis during MNV and that WTAP may participate in the formation of MNV through the wingless-related integration site (Wnt) pathway. WTAP may be a potential target for MNV treatment.
Collapse
Affiliation(s)
- Qingyun Gong
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Liting Hu
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Guibo Liu
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaoni Yin
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaoran Zhao
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qinghua Li
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ying Li
- Department of Ophthalmology, Linyi People's Hospital, Shandong, China
| | - Yibin Sun
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yuzheng Zhou
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Chunyan Guo
- Department of Ophthalmology, Gansu Province, China
| | - Zhaodong Du
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
9
|
Chen H, Jiao Y, Lin C, Fan W, Li L, Li B, Li L, Zeng X, Li Z, Wei H, Zhang Y, Zhou B, Chen C, Ye J, Yang M. Thrombopoietin improves the functions of bone marrow endothelial progenitor cells via METTL16/Akt signalling of haematological patients with chemotherapy-induced thrombocytopenia. Br J Haematol 2024; 205:1532-1545. [PMID: 39189039 DOI: 10.1111/bjh.19722] [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: 03/31/2024] [Accepted: 08/12/2024] [Indexed: 08/28/2024]
Abstract
Bone marrow endothelial progenitor cells (BM EPCs) are crucial in supporting haematopoietic regeneration, while the BM EPCs of haematological patients with chemotherapy-induced thrombocytopenia (CIT) are unavoidably damaged. Therefore, the present study aimed to examine the effect of thrombopoietin (TPO) on the recovery of BM EPCs of CIT patients and to identify the underlying mechanisms. The cell functions were determined by 1,1'-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate (Dil)-acetylated low-density lipoprotein (Dil-Ac-LDL) uptake and fluorescein isothiocyanate (FITC)-labeled Ulex europaeus agglutinin-I (FITC-UEA-I) binding assay, as well as proliferation, migration and tube formation experiments. Endothelial cells were transfected with METTL16 lentivirus, followed by methylated RNA immunoprecipitation sequencing. Zebrafish with vascular defect was used as the in vivo model. TPO significantly improved the quantity and functions of BM EPCs from CIT patients in vitro and restored the subintestinal vein area of zebrafish with vascular defect in vivo. Mechanically, TPO enhanced the BM EPC functions through Akt signal mediated by METTL16, which was downregulated in BM EPCs of CIT patients and involved in the regulation of endothelial functions. The present study demonstrates that TPO improves the recovery of BM EPCs from CIT patients with haematological malignancies via METTL16/Akt signalling, which provides new insights into the role of TPO in treating CIT in addition to direct megakaryopoiesis.
Collapse
Affiliation(s)
- Hui Chen
- The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, P.R. China
- Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, Shenzhen, Guangdong, P.R. China
| | - Yingying Jiao
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, P.R. China
| | - Chao Lin
- The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, P.R. China
| | - Wenxuan Fan
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, P.R. China
| | - Lindi Li
- The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, P.R. China
| | - Bo Li
- The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, P.R. China
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, Shenzhen, Guangdong, P.R. China
| | - Liang Li
- The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, P.R. China
- Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, Shenzhen, Guangdong, P.R. China
| | - Xiaoyuan Zeng
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, P.R. China
| | - Zongpeng Li
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, P.R. China
| | - Hongfa Wei
- The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, P.R. China
| | - Yuming Zhang
- Department of Hematology, Hematology Research Institute, Affiliated Hospital of Guangdong Medical University (GDMU), Zhanjiang, China
| | - Benjie Zhou
- The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, P.R. China
- Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, Shenzhen, Guangdong, P.R. China
| | - Chun Chen
- The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, P.R. China
| | - Jieyu Ye
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, P.R. China
| | - Mo Yang
- The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, P.R. China
- Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, Shenzhen, Guangdong, P.R. China
- Department of Hematology, Hematology Research Institute, Affiliated Hospital of Guangdong Medical University (GDMU), Zhanjiang, China
| |
Collapse
|
10
|
Wang Y, Zou J, Zhou H. N6-methyladenine RNA methylation epigenetic modification and diabetic microvascular complications. Front Endocrinol (Lausanne) 2024; 15:1462146. [PMID: 39296713 PMCID: PMC11408340 DOI: 10.3389/fendo.2024.1462146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 08/20/2024] [Indexed: 09/21/2024] Open
Abstract
N6-methyladensine (m6A) has been identified as the best-characterized and the most abundant mRNA modification in eukaryotes. It can be dynamically regulated, removed, and recognized by its specific cellular components (respectively called "writers," "erasers," "readers") and have become a hot research field in a variety of biological processes and diseases. Currently, the underlying molecular mechanisms of m6A epigenetic modification in diabetes mellitus (DM) and diabetic microvascular complications have not been extensively clarified. In this review, we focus on the effects and possible mechanisms of m6A as possible potential biomarkers and therapeutic targets in the treatment of DM and diabetic microvascular complications.
Collapse
Affiliation(s)
- Yuanyuan Wang
- Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jiayun Zou
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Hua Zhou
- Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
11
|
Sun H, Gao Y, Ma X, Deng Y, Bi L, Li L. Mechanism and application of feedback loops formed by mechanotransduction and histone modifications. Genes Dis 2024; 11:101061. [PMID: 39071110 PMCID: PMC11282412 DOI: 10.1016/j.gendis.2023.06.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 03/24/2023] [Accepted: 06/27/2023] [Indexed: 07/30/2024] Open
Abstract
Mechanical stimulation is the key physical factor in cell environment. Mechanotransduction acts as a fundamental regulator of cell behavior, regulating cell proliferation, differentiation, apoptosis, and exhibiting specific signature alterations during the pathological process. As research continues, the role of epigenetic science in mechanotransduction is attracting attention. However, the molecular mechanism of the synergistic effect between mechanotransduction and epigenetics in physiological and pathological processes has not been clarified. We focus on how histone modifications, as important components of epigenetics, are coordinated with multiple signaling pathways to control cell fate and disease progression. Specifically, we propose that histone modifications can form regulatory feedback loops with signaling pathways, that is, histone modifications can not only serve as downstream regulators of signaling pathways for target gene transcription but also provide feedback to regulate signaling pathways. Mechanotransduction and epigenetic changes could be potential markers and therapeutic targets in clinical practice.
Collapse
Affiliation(s)
- Han Sun
- Department of Hematology and Oncology, China-Japan Union Hospital, Jilin University, Changchun, Jilin 130021, China
| | - Yafang Gao
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, China
| | - Xinyu Ma
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, China
| | - Yizhou Deng
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, China
| | - Lintao Bi
- Department of Hematology and Oncology, China-Japan Union Hospital, Jilin University, Changchun, Jilin 130021, China
| | - Lisha Li
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, China
| |
Collapse
|
12
|
Chen K, Li WD, Li XQ. The role of m6A in angiogenesis and vascular diseases. iScience 2024; 27:110082. [PMID: 39055919 PMCID: PMC11269316 DOI: 10.1016/j.isci.2024.110082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/28/2024] Open
Abstract
Angiogenesis, whether physiological or pathological, plays a pivotal role in various physiological and disease conditions. This intricate process relies on a complex and meticulously orchestrated signal transduction network that connects endothelial cells, their associated parietal cells (VSMCs and pericytes), and various other cell types, including immune cells. Given the significance of m6A and its connection to angiogenesis and vascular disease, researchers must adopt a comprehensive and ongoing approach to their investigations. This study aims to ascertain whether a common key mechanism of m6A exists in angiogenesis and vascular diseases and to elucidate the potential application of m6A in treating vascular diseases.
Collapse
Affiliation(s)
- Ke Chen
- Department of Vascular Surgery, The Drum Tower Hospital Affiliated to Medical School of Nanjing University, Nanjing, China
| | - Wen-Dong Li
- Department of Vascular Surgery, The Drum Tower Hospital Affiliated to Medical School of Nanjing University, Nanjing, China
| | - Xiao-Qiang Li
- Department of Vascular Surgery, The Drum Tower Hospital Affiliated to Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
13
|
Liu J, Chai XX, Qiu XR, Sun WJ, Tian YL, Guo WH, Yin DC, Zhang CY. Type X collagen knockdown inactivate ITGB1/PI3K/AKT to suppress chronic unpredictable mild stress-stimulated triple-negative breast cancer progression. Int J Biol Macromol 2024; 273:133074. [PMID: 38866293 DOI: 10.1016/j.ijbiomac.2024.133074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 05/19/2024] [Accepted: 06/08/2024] [Indexed: 06/14/2024]
Abstract
Triple-negative breast cancer (TNBC) is the most malignant subtype of breast cancer, has a poor prognosis and limited access to efficient targeted treatments. Chronic unpredictable mild stress (CUMS) is highly risk factor for TNBC occurrence and development. Type X collagen (COL10A1), a crucial protein component of the extracellular matrix, ranks second among all aberrantly expressed genes in TNBC, and it is significantly up-regulated under CUMS. Nevertheless, the impact of CUMS and COL10A1 on TNBC, along with the underlying mechanisms are still unclear. In this research, we studied the effect of CUMS-induced norepinephrine (NE) elevation on TNBC, and uncovered that it notably enhanced TNBC cell proliferation, migration, and invasion in vitro, and also fostering tumor growth and lung metastasis in vivo. Additionally, our investigation found that COL10A1 directly interacted with integrin subunit beta 1 (ITGB1), then activates the downstream PI3K/AKT signaling pathway, thereby promoting TNBC growth and metastasis, while it was reversed by knocking down of COL10A1 or ITGB1. Our study demonstrated that the TNBC could respond to CUMS, and advocate for COL10A1 as a pivotal therapeutic target in TNBC treatment.
Collapse
Affiliation(s)
- Jie Liu
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710000, Shaanxi, PR China
| | - Xiao-Xia Chai
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710000, Shaanxi, PR China
| | - Xiao-Rong Qiu
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710000, Shaanxi, PR China
| | - Wen-Jun Sun
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710000, Shaanxi, PR China
| | - Yi-Le Tian
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710000, Shaanxi, PR China
| | - Wei-Hong Guo
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710000, Shaanxi, PR China
| | - Da-Chuan Yin
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710000, Shaanxi, PR China.
| | - Chen-Yan Zhang
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710000, Shaanxi, PR China; Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen 518063, China.
| |
Collapse
|
14
|
Luo D, Tang H, Tan L, Zhang L, Wang L, Cheng Q, Lei X, Wu J. lncRNA JPX Promotes Tumor Progression by Interacting with and Destabilizing YTHDF2 in Cutaneous Melanoma. Mol Cancer Res 2024; 22:524-537. [PMID: 38441563 DOI: 10.1158/1541-7786.mcr-23-0701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 01/18/2024] [Accepted: 03/01/2024] [Indexed: 06/05/2024]
Abstract
Aberrant long noncoding RNAs just proximal to Xist (lncRNA JPX) expression levels have been detected in multiple tumors. However, whether JPX is involved in melanoma progression remains unclear. Our study showed that JPX expression is significantly increased in melanoma tissues and cell lines. To clarify the effect of JPX on cutaneous melanoma, we successfully generated JPX-overexpressing or JPX-knockdown A375 and A2058 cells. CCK-8, colony formation EdU, Transwell, and cell-cycle phase assays were performed, and subcutaneously implanted tumor models were used to determine the function of JPX in cutaneous melanoma. The results showed that JPX knockdown reduced the proliferation and migration of malignant melanoma cells both in vitro and in vivo. To further elucidate the molecular mechanism of JPX-induced cutaneous melanoma deterioration, we performed RNA pull-down, RNA immunoprecipitation, coimmunoprecipitation, Western blot, and RNA-sequence analyses. JPX can directly interact with YTHDF2 and impede the protection of YTHDF2 from ubiquitin-specific protease 10 (USP10), which promotes its deubiquitination. Thus, JPX decreases protein stability and promotes the degradation of YTHDF2, thereby stabilizing BMP2 mRNA and activating AKT phosphorylation. Overall, our study revealed a novel effect of JPX on YTHDF2 ubiquitination, suggesting the possibility of blocking the JPX/USP10/YTHDF2/BMP2 axis as a prospective therapeutic approach for cutaneous melanoma. IMPLICATIONS This study highlights the ubiquitination effect of USP10 and JPX on YTHDF2 in cutaneous melanoma, and proposes that the JPX/USP10/YTHDF2/BMP2 axis may be a prospective therapeutic target for cutaneous melanoma.
Collapse
Affiliation(s)
- Dan Luo
- Department of Dermatology, Daping Hospital, Army Medical University, Chongqing, China
| | - Hui Tang
- Department of Dermatology, Daping Hospital, Army Medical University, Chongqing, China
| | - Liuchang Tan
- Department of Plastic and Cosmetic Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Long Zhang
- Department of Dermatology, Daping Hospital, Army Medical University, Chongqing, China
| | - Lei Wang
- Department of Dermatology, Daping Hospital, Army Medical University, Chongqing, China
| | - Qionghui Cheng
- Department of Dermatology, Daping Hospital, Army Medical University, Chongqing, China
| | - Xia Lei
- Department of Dermatology, Daping Hospital, Army Medical University, Chongqing, China
| | - Jinjin Wu
- Department of Dermatology, Daping Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
15
|
Lin P, Cao W, Chen X, Zhang N, Xing Y, Yang N. Role of mRNA-binding proteins in retinal neovascularization. Exp Eye Res 2024; 242:109870. [PMID: 38514023 DOI: 10.1016/j.exer.2024.109870] [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: 01/27/2024] [Revised: 03/06/2024] [Accepted: 03/18/2024] [Indexed: 03/23/2024]
Abstract
Retinal neovascularization (RNV) is a pathological process that primarily occurs in diabetic retinopathy, retinopathy of prematurity, and retinal vein occlusion. It is a common yet debilitating clinical condition that culminates in blindness. Urgent efforts are required to explore more efficient and less limiting therapeutic strategies. Key RNA-binding proteins (RBPs), crucial for post-transcriptional regulation of gene expression by binding to RNAs, are closely correlated with RNV development. RBP-RNA interactions are altered during RNV. Here, we briefly review the characteristics and functions of RBPs, and the mechanism of RNV. Then, we present insights into the role of the regulatory network of RBPs in RNV. HuR, eIF4E, LIN28B, SRSF1, METTL3, YTHDF1, Gal-1, HIWI1, and ZFR accelerate RNV progression, whereas YTHDF2 and hnRNPA2B1 hinder it. The mechanisms elucidated in this review provide a reference to guide the design of therapeutic strategies to reverse abnormal processes.
Collapse
Affiliation(s)
- Pei Lin
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Jiefang Road #238, Wuhan, 430060, Hubei, China.
| | - Wenye Cao
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Jiefang Road #238, Wuhan, 430060, Hubei, China.
| | - Xuemei Chen
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Jiefang Road #238, Wuhan, 430060, Hubei, China.
| | - Ningzhi Zhang
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Jiefang Road #238, Wuhan, 430060, Hubei, China.
| | - Yiqiao Xing
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Jiefang Road #238, Wuhan, 430060, Hubei, China; Department of Ophthalmology, Aier Eye Hospital of Wuhan University, Hubei, China.
| | - Ning Yang
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Jiefang Road #238, Wuhan, 430060, Hubei, China.
| |
Collapse
|
16
|
Jin Q, Liu T, Ma F, Fu T, Yang L, Mao H, Wang Y, Peng L, Li P, Zhan Y. Roles of Sirt1 and its modulators in diabetic microangiopathy: A review. Int J Biol Macromol 2024; 264:130761. [PMID: 38467213 DOI: 10.1016/j.ijbiomac.2024.130761] [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: 12/27/2023] [Revised: 02/03/2024] [Accepted: 03/07/2024] [Indexed: 03/13/2024]
Abstract
Diabetic vascular complications include diabetic macroangiopathy and diabetic microangiopathy. Diabetic microangiopathy is characterised by impaired microvascular endothelial function, basement membrane thickening, and microthrombosis, which may promote renal, ocular, cardiac, and peripheral system damage in diabetic patients. Therefore, new preventive and therapeutic strategies are urgently required. Sirt1, a member of the nicotinamide adenine dinucleotide-dependent histone deacetylase class III family, regulates different organ growth and development, oxidative stress, mitochondrial function, metabolism, inflammation, and aging. Sirt1 is downregulated in vascular injury and microangiopathy. Moreover, its expression and distribution in different organs correlate with age and play critical regulatory roles in oxidative stress and inflammation. This review introduces the background of diabetic microangiopathy and the main functions of Sirt1. Then, the relationship between Sirt1 and different diabetic microangiopathies and the regulatory roles mediated by different cells are described. Finally, we summarize the modulators that target Sirt1 to ameliorate diabetic microangiopathy as an essential preventive and therapeutic measure for diabetic microangiopathy. In conclusion, targeting Sirt1 may be a new therapeutic strategy for diabetic microangiopathy.
Collapse
Affiliation(s)
- Qi Jin
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Tongtong Liu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Fang Ma
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Tongfei Fu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Liping Yang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Huimin Mao
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuyang Wang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Liang Peng
- China-Japan Friendship Hospital, Institute of Clinical Medical Sciences, Beijing, China.
| | - Ping Li
- China-Japan Friendship Hospital, Institute of Clinical Medical Sciences, Beijing, China.
| | - Yongli Zhan
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
17
|
Li B, Xuan H, Yin Y, Wu S, Du L. The N 6-methyladenosine modification in pathologic angiogenesis. Life Sci 2024; 339:122417. [PMID: 38244915 DOI: 10.1016/j.lfs.2024.122417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 01/03/2024] [Accepted: 01/07/2024] [Indexed: 01/22/2024]
Abstract
The vascular system is a vital circulatory network in the human body that plays a critical role in almost all physiological processes. The production of blood vessels in the body is a significant area of interest for researchers seeking to improve their understanding of vascular function and maintain normal vascular operation. However, an excessive or insufficient vascular regeneration process may lead to the development of various ailments such as cancer, eye diseases, and ischemic diseases. Recent preclinical and clinical studies have revealed new molecular targets and principles that may enhance the therapeutic effect of anti-angiogenic strategies. A thorough comprehension of the mechanism responsible for the abnormal vascular growth in disease processes can enable researchers to better target and effectively suppress or treat the disease. N6-methyladenosine (m6A), a common RNA methylation modification method, has emerged as a crucial regulator of various diseases by modulating vascular development. In this review, we will cover how m6A regulates various vascular-related diseases, such as cancer, ocular diseases, neurological diseases, ischemic diseases, emphasizing the mechanism of m6A methylation regulators on angiogenesis during pathological process.
Collapse
Affiliation(s)
- Bin Li
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Hanqin Xuan
- Department of Pathology, the First Affiliated Hospital of Soochow University, Jiangsu, China
| | - Yuye Yin
- College of Bioscience and Biotechnology, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Shusheng Wu
- Department of Neurology, Affiliated Hospital of Yangzhou University, Jiangsu, China.
| | - Longfei Du
- Department of Laboratory Medicine, Affiliated Hospital of Yangzhou University, Yangzhou, Jiangsu, China.
| |
Collapse
|
18
|
Chen X, Wang Y, Wang JN, Zhang YC, Zhang YR, Sun RX, Qin B, Dai YX, Zhu HJ, Zhao JX, Zhang WW, Ji JD, Yuan ST, Shen QD, Liu QH. Lactylation-driven FTO targets CDK2 to aggravate microvascular anomalies in diabetic retinopathy. EMBO Mol Med 2024; 16:294-318. [PMID: 38297099 PMCID: PMC10897304 DOI: 10.1038/s44321-024-00025-1] [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: 06/01/2023] [Revised: 12/10/2023] [Accepted: 01/02/2024] [Indexed: 02/02/2024] Open
Abstract
Diabetic retinopathy (DR) is a leading cause of irreversible vision loss in working-age populations. Fat mass and obesity-associated protein (FTO) is an N6-methyladenosine (m6A) demethylase that demethylates RNAs involved in energy homeostasis, though its influence on DR is not well studied. Herein, we detected elevated FTO expression in vitreous fibrovascular membranes of patients with proliferative DR. FTO promoted cell cycle progression and tip cell formation of endothelial cells (ECs) to facilitate angiogenesis in vitro, in mice, and in zebrafish. FTO also regulated EC-pericyte crosstalk to trigger diabetic microvascular leakage, and mediated EC-microglia interactions to induce retinal inflammation and neurodegeneration in vivo and in vitro. Mechanistically, FTO affected EC features via modulating CDK2 mRNA stability in an m6A-YTHDF2-dependent manner. FTO up-regulation under diabetic conditions was driven by lactate-mediated histone lactylation. FB23-2, an inhibitor to FTO's m6A demethylase activity, suppressed angiogenic phenotypes in vitro. To allow for systemic administration, we developed a nanoplatform encapsulating FB23-2 and confirmed its targeting and therapeutic efficiency in mice. Collectively, our study demonstrates that FTO is important for EC function and retinal homeostasis in DR, and warrants further investigation as a therapeutic target for DR patients.
Collapse
Affiliation(s)
- Xue Chen
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China.
| | - Ying Wang
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Jia-Nan Wang
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Yi-Chen Zhang
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Ye-Ran Zhang
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Ru-Xu Sun
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Bing Qin
- Department of Ophthalmology, The Affiliated Suqian First People's Hospital of Nanjing Medical University, Suqian, China
| | - Yuan-Xin Dai
- Department of Polymer Science and Engineering and Key Laboratory of High-Performance Polymer Materials and Technology of MOE, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, China
| | - Hong-Jing Zhu
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Jin-Xiang Zhao
- Department of Ophthalmology, The Affiliated Suqian First People's Hospital of Nanjing Medical University, Suqian, China
| | - Wei-Wei Zhang
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Jiang-Dong Ji
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Song-Tao Yuan
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Qun-Dong Shen
- Department of Polymer Science and Engineering and Key Laboratory of High-Performance Polymer Materials and Technology of MOE, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, China
| | - Qing-Huai Liu
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
19
|
Su C, Mo J, Dong S, Liao Z, Zhang B, Zhu P. Integrinβ-1 in disorders and cancers: molecular mechanisms and therapeutic targets. Cell Commun Signal 2024; 22:71. [PMID: 38279122 PMCID: PMC10811905 DOI: 10.1186/s12964-023-01338-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 09/27/2023] [Indexed: 01/28/2024] Open
Abstract
Integrinβ-1 (ITGB1) is a crucial member of the transmembrane glycoprotein signaling receptor family and is also central to the integrin family. It forms heterodimers with other ligands, participates in intracellular signaling and controls a variety of cellular processes, such as angiogenesis and the growth of neurons; because of its role in bidirectional signaling regulation both inside and outside the membrane, ITGB1 must interact with a multitude of substances, so a variety of interfering factors can affect ITGB1 and lead to changes in its function. Over the past 20 years, many studies have confirmed a clear causal relationship between ITGB1 dysregulation and cancer development and progression in a wide range of benign diseases and solid tumor types, which may imply that ITGB1 is a prognostic biomarker and a therapeutic target for cancer treatment that warrants further investigation. This review summarizes the biological roles of ITGB1 in benign diseases and cancers, and compiles the current status of ITGB1 function and therapy in various aspects of tumorigenesis and progression. Finally, future research directions and application prospects of ITGB1 are suggested. Video Abstract.
Collapse
Affiliation(s)
- Chen Su
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, People's Republic of China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, People's Republic of China
| | - Jie Mo
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, People's Republic of China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, People's Republic of China
| | - Shuilin Dong
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, People's Republic of China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, People's Republic of China
| | - Zhibin Liao
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, People's Republic of China.
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, People's Republic of China.
| | - Bixiang Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, People's Republic of China.
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, People's Republic of China.
- Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, Hubei, People's Republic of China.
- Key Laboratory of Organ Transplantation, National Health Commission, Wuhan, Hubei, People's Republic of China.
- Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, Hubei, People's Republic of China.
| | - Peng Zhu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, People's Republic of China.
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, People's Republic of China.
- Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, Hubei, People's Republic of China.
- Key Laboratory of Organ Transplantation, National Health Commission, Wuhan, Hubei, People's Republic of China.
- Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, Hubei, People's Republic of China.
| |
Collapse
|
20
|
Li B, Wang Z, Zhou H, Zou J, Yoshida S, Zhou Y. N6-methyladenosine methylation in ophthalmic diseases: From mechanisms to potential applications. Heliyon 2024; 10:e23668. [PMID: 38192819 PMCID: PMC10772099 DOI: 10.1016/j.heliyon.2023.e23668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 11/23/2023] [Accepted: 12/09/2023] [Indexed: 01/10/2024] Open
Abstract
N6-methyladenosine (m6A) modification, as the most common modification method in eukaryotes, is widely involved in numerous physiological and pathological processes, such as embryonic development, malignancy, immune regulation, and premature aging. Under pathological conditions of ocular diseases, changes in m6A modification and its metabolism can be detected in aqueous and vitreous humor. At the same time, an increasing number of studies showed that m6A modification is involved in the normal development of eye structures and the occurrence and progress of many ophthalmic diseases, especially ocular neovascular diseases, such as diabetic retinopathy, age-related macular degeneration, and melanoma. In this review, we summarized the latest progress regarding m6A modification in ophthalmic diseases, changes in m6A modification-related enzymes in various pathological states and their upstream and downstream regulatory networks, provided new prospects for m6A modification in ophthalmic diseases and new ideas for clinical diagnosis and treatment.
Collapse
Affiliation(s)
- Bingyan Li
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China
- National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Zicong Wang
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China
- National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Haixiang Zhou
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China
- National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Jingling Zou
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China
- National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Shigeo Yoshida
- Department of Ophthalmology, Kurume University School of Medicine, Kurume, Fukuoka, 830-0011, Japan
| | - Yedi Zhou
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China
- National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| |
Collapse
|
21
|
Yu W, Yang B, Xu S, Gao Y, Huang Y, Wang Z. Diabetic Retinopathy and Cardiovascular Disease: A Literature Review. Diabetes Metab Syndr Obes 2023; 16:4247-4261. [PMID: 38164419 PMCID: PMC10758178 DOI: 10.2147/dmso.s438111] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 12/21/2023] [Indexed: 01/03/2024] Open
Abstract
Diabetic complications can be divided into macrovascular complications such as cardiovascular disease and cerebrovascular disease and microvascular complications such as diabetic retinopathy, diabetic nephropathy and diabetic neuropathy. Among them, cardiovascular disease (CVD) is an important cause of death in diabetic patients. Diabetes retinopathy (DR) is one of the main reasons for the increasing disability rate of diabetes. In recent years, some studies have found that because DR and CVD have a common pathophysiological basis, the occurrence of DR and CVD are inseparable, and to a certain extent, DR can predict the occurrence of CVD. With the development of technology, the fundus parameters of DR can be quantitatively analyzed as an independent risk factor of CVD. In addition, the cytokines related to DR can also be used for early screening of DR. Although many advances have been made in the treatment of CVD, its situation of prevention and treatment is still not optimistic. This review hopes to discuss the feasibility of DR in predicting CVD from the common pathophysiological mechanism of DR and CVD, the new progress of diagnostic techniques for DR, and the biomarkers for early screening of DR.
Collapse
Affiliation(s)
- Wenhua Yu
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, People’s Republic of China
| | - Bo Yang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, People’s Republic of China
| | - Siting Xu
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, People’s Republic of China
| | - Yun Gao
- Department of Pathology, Affiliated Hospital of Jiangsu University, Zhenjiang, People’s Republic of China
| | - Yan Huang
- Department of Ophthalmology, Affiliated Hospital of Jiangsu University, Zhenjiang, People’s Republic of China
| | - Zhongqun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, People’s Republic of China
| |
Collapse
|
22
|
Ren Y, Li Z, Li J, Liang R, Wang Z, Bai Y, Yang Y, Tang Q, Fu Y, Zhang X, Zhang Y, Yu Y, Xiong Y. m 6 A mRNA methylation: Biological features, mechanisms, and therapeutic potentials in type 2 diabetes mellitus. Obes Rev 2023; 24:e13639. [PMID: 37732463 DOI: 10.1111/obr.13639] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/10/2023] [Accepted: 08/27/2023] [Indexed: 09/22/2023]
Abstract
As the most common internal post-transcriptional RNA modification in eukaryotic cells, N6-methyladenosine (m6 A) performs a dynamic and reversible role in a variety of biological processes mediated by methyltransferases (writers), demethylases (erasers), and m6 A binding proteins (readers). M6 A methylation enables transcriptome conversion in different signals that regulate various physiological activities and organ development. Over the past few years, emerging studies have identified that mRNA m6 A regulators defect in β-cell leads to abnormal regulation of the target mRNAs, thereby resulting in β-cell dysfunction and loss of β-cell identity and mass, which are strongly associated with type 2 diabetes mellitus (T2DM) pathogenesis. Also, mRNA m6 A modification has been implicated with insulin resistance in muscles, fat, and liver cells/tissues. In this review, we elaborate on the biological features of m6 A methylation; provide a comprehensive overview of the underlying mechanisms that how it controls β-cell function, identity, and mass as well as insulin resistance; highlight its connections to glucose metabolism and lipid metabolism linking to T2DM; and further discuss its role in diabetes complications and its therapeutic potentials for T2DM diagnosis and treatment.
Collapse
Affiliation(s)
- Yuanyuan Ren
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Zi Li
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Jiaoyu Li
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Rui Liang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Zhen Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Yiduo Bai
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Yafang Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Qian Tang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Yaolei Fu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Xiaobo Zhang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Yu Zhang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Yi Yu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
- School of Medicine, Northwest University, Xi'an, Shaanxi, China
| | - Yuyan Xiong
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
- School of Medicine, Northwest University, Xi'an, Shaanxi, China
| |
Collapse
|
23
|
Zhuang A, Gu X, Ge T, Wang S, Ge S, Chai P, Jia R, Fan X. Targeting histone deacetylase suppresses tumor growth through eliciting METTL14-modified m 6 A RNA methylation in ocular melanoma. Cancer Commun (Lond) 2023; 43:1185-1206. [PMID: 37466203 PMCID: PMC10631484 DOI: 10.1002/cac2.12471] [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: 11/13/2022] [Revised: 03/31/2023] [Accepted: 07/13/2023] [Indexed: 07/20/2023] Open
Abstract
BACKGROUND Diversified histone deacetylation inhibitors (HDACis) have demonstrated encouraging outcomes in multiple malignancies. N6-methyladenine (m6 A) is the most prevalent messenger RNA modification that plays an essential role in the regulation of tumorigenesis. Howbeit, an in-depth understanding of the crosstalk between histone acetylation and m6 A RNA modifications remains enigmatic. This study aimed to explore the role of histone acetylation and m6 A modifications in the regulation of tumorigenesis of ocular melanoma. METHODS Histone modification inhibitor screening was used to explore the effects of HDACis on ocular melanoma cells. Dot blot assay was used to detect the global m6 A RNA modification level. Multi-omics assays, including RNA-sequencing, cleavage under targets and tagmentation, single-cell sequencing, methylated RNA immunoprecipitation-sequencing (meRIP-seq), and m6 A individual nucleotide resolution cross-linking and immunoprecipitation-sequencing (miCLIP-seq), were performed to reveal the mechanisms of HDACis on methyltransferase-like 14 (METTL14) and FAT tumor suppressor homolog 4 (FAT4) in ocular melanoma. Quantitative real-time polymerase chain reaction (qPCR), western blotting, and immunofluorescent staining were applied to detect the expression of METTL14 and FAT4 in ocular melanoma cells and tissues. Cell models and orthotopic xenograft models were established to determine the roles of METTL14 and FAT4 in the growth of ocular melanoma. RNA-binding protein immunoprecipitation-qPCR, meRIP-seq, miCLIP-seq, and RNA stability assay were adopted to investigate the mechanism by which m6 A levels of FAT4 were affected. RESULTS First, we found that ocular melanoma cells presented vulnerability towards HDACis. HDACis triggered the elevation of m6 A RNA modification in ocular melanoma. Further studies revealed that METTL14 served as a downstream candidate for HDACis. METTL14 was silenced by the hypo-histone acetylation status, whereas HDACi restored the normal histone acetylation level of METTL14, thereby inducing its expression. Subsequently, METTL14 served as a tumor suppressor by promoting the expression of FAT4, a tumor suppressor, in a m6 A-YTH N6-methyladenosine RNA-binding protein 1-dependent manner. Taken together, we found that HDACi restored the histone acetylation level of METTL14 and subsequently elicited METTL14-mediated m6 A modification in tumorigenesis. CONCLUSIONS These results demonstrate that HDACis exert anti-cancer effects by orchestrating m6 A modification, which unveiling a "histone-RNA crosstalk" of the HDAC/METTL14/FAT4 epigenetic cascade in ocular melanoma.
Collapse
Affiliation(s)
- Ai Zhuang
- Department of OphthalmologyShanghai Ninth People's HospitalShanghai JiaoTong University School of MedicineShanghaiP. R. China
- Shanghai Key Laboratory of Orbital Diseases and Ocular OncologyShanghaiP. R. China
| | - Xiang Gu
- Department of OphthalmologyShanghai Ninth People's HospitalShanghai JiaoTong University School of MedicineShanghaiP. R. China
- Shanghai Key Laboratory of Orbital Diseases and Ocular OncologyShanghaiP. R. China
| | - Tongxin Ge
- Department of OphthalmologyShanghai Ninth People's HospitalShanghai JiaoTong University School of MedicineShanghaiP. R. China
- Shanghai Key Laboratory of Orbital Diseases and Ocular OncologyShanghaiP. R. China
| | - Shaoyun Wang
- Department of OphthalmologyShanghai Ninth People's HospitalShanghai JiaoTong University School of MedicineShanghaiP. R. China
- Shanghai Key Laboratory of Orbital Diseases and Ocular OncologyShanghaiP. R. China
| | - Shengfang Ge
- Department of OphthalmologyShanghai Ninth People's HospitalShanghai JiaoTong University School of MedicineShanghaiP. R. China
- Shanghai Key Laboratory of Orbital Diseases and Ocular OncologyShanghaiP. R. China
| | - Peiwei Chai
- Department of OphthalmologyShanghai Ninth People's HospitalShanghai JiaoTong University School of MedicineShanghaiP. R. China
- Shanghai Key Laboratory of Orbital Diseases and Ocular OncologyShanghaiP. R. China
| | - Renbing Jia
- Department of OphthalmologyShanghai Ninth People's HospitalShanghai JiaoTong University School of MedicineShanghaiP. R. China
- Shanghai Key Laboratory of Orbital Diseases and Ocular OncologyShanghaiP. R. China
| | - Xianqun Fan
- Department of OphthalmologyShanghai Ninth People's HospitalShanghai JiaoTong University School of MedicineShanghaiP. R. China
- Shanghai Key Laboratory of Orbital Diseases and Ocular OncologyShanghaiP. R. China
| |
Collapse
|
24
|
Lin LC, Liu ZY, Yang JJ, Zhao JY, Tao H. m6A epitranscriptomic modification in diabetic microvascular complications. Trends Pharmacol Sci 2023; 44:S0165-6147(23)00215-8. [PMID: 39492320 DOI: 10.1016/j.tips.2023.09.013] [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: 08/24/2023] [Revised: 09/21/2023] [Accepted: 09/29/2023] [Indexed: 11/05/2024]
Abstract
N6-methyladenosine (m6A) modifications are modulated by m6A methyltransferases, m6A demethylases, and m6A-binding proteins. The dynamic and reversible patterns of m6A modification control cell fate programming by regulating RNA splicing, translation, and decay. Emerging evidence demonstrates that m6A modification of coding and noncoding RNAs exerts crucial effects that influence the pathogenesis of diabetic microvascular complications that include diabetic cardiomyopathy, diabetic nephropathy, diabetic retinopathy, diabetic neuropathy, and diabetic dermatosis. In this review, we summarize the roles of m6A modification and m6A modification-related enzymes in diabetic microvascular complications and discuss potential m6A modification-related enzyme-targeting therapeutic strategies.
Collapse
Affiliation(s)
- Li-Chan Lin
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Zhi-Yan Liu
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Jing-Jing Yang
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.
| | - Jian-Yuan Zhao
- Institute for Developmental and Regenerative Cardiovascular Medicine, MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| | - Hui Tao
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China; Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.
| |
Collapse
|
25
|
Sun RX, Zhu HJ, Zhang YR, Wang JN, Wang Y, Cao QC, Ji JD, Jiang C, Yuan ST, Chen X, Liu QH. ALKBH5 causes retinal pigment epithelium anomalies and choroidal neovascularization in age-related macular degeneration via the AKT/mTOR pathway. Cell Rep 2023; 42:112779. [PMID: 37436898 DOI: 10.1016/j.celrep.2023.112779] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 05/24/2023] [Accepted: 06/25/2023] [Indexed: 07/14/2023] Open
Abstract
Retinal pigment epithelium (RPE) dysfunction and choroidal neovascularization (CNV) are predominant features of age-related macular degeneration (AMD), with an unclear mechanism. Herein, we show that RNA demethylase α-ketoglutarate-dependent dioxygenase alkB homolog 5 (ALKBH5) is up-regulated in AMD. In RPE cells, ALKBH5 overexpression associates with depolarization, oxidative stress, disturbed autophagy, irregular lipid homeostasis, and elevated VEGF-A secretion, which subsequently promotes proliferation, migration, and tube formation of vascular endothelial cells. Consistently, ALKBH5 overexpression in mice RPE correlates with various pathological phenotypes, including visual impairments, RPE anomalies, choroidal neovascularization (CNV), and interrupted retinal homeostasis. Mechanistically, ALKBH5 regulates retinal features through its demethylation activity. It targets PIK3C2B and regulates the AKT/mTOR signaling pathway with YTHDF2 as the N6-methyladenosine reader. IOX1, an ALKBH5 inhibitor, suppresses hypoxia-induced RPE dysfunction and CNV progression. Collectively, we demonstrate that ALKBH5 induces RPE dysfunction and CNV progression in AMD via PIK3C2B-mediated activation of the AKT/mTOR pathway. Pharmacological inhibitors of ALKBH5, like IOX1, are promising therapeutic options for AMD.
Collapse
Affiliation(s)
- Ru-Xu Sun
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210029, China
| | - Hong-Jing Zhu
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210029, China
| | - Ye-Ran Zhang
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210029, China
| | - Jia-Nan Wang
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210029, China
| | - Ying Wang
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210029, China
| | - Qiu-Chen Cao
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210029, China
| | - Jiang-Dong Ji
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210029, China
| | - Chao Jiang
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210029, China
| | - Song-Tao Yuan
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210029, China.
| | - Xue Chen
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210029, China.
| | - Qing-Huai Liu
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210029, China.
| |
Collapse
|
26
|
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
|
27
|
Liu DD, Zhang CY, Zhang JT, Gu LM, Xu GT, Zhang JF. Epigenetic modifications and metabolic memory in diabetic retinopathy: beyond the surface. Neural Regen Res 2023; 18:1441-1449. [PMID: 36571340 PMCID: PMC10075108 DOI: 10.4103/1673-5374.361536] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 10/10/2022] [Accepted: 10/26/2022] [Indexed: 11/19/2022] Open
Abstract
Epigenetics focuses on DNA methylation, histone modification, chromatin remodeling, noncoding RNAs, and other gene regulation mechanisms beyond the DNA sequence. In the past decade, epigenetic modifications have drawn more attention as they participate in the development and progression of diabetic retinopathy despite tight control of glucose levels. The underlying mechanisms of epigenetic modifications in diabetic retinopathy still urgently need to be elucidated. The diabetic condition facilitates epigenetic changes and influences target gene expression. In this review, we summarize the involvement of epigenetic modifications and metabolic memory in the development and progression of diabetic retinopathy and propose novel insights into the treatment of diabetic retinopathy.
Collapse
Affiliation(s)
- Dan-Dan Liu
- Department of Ophthalmology of Tongji Hospital, Tongji Eye Institute, Department of Regenerative Medicine, and Department of Pharmacology, Tongji University School of Medicine, Shanghai, China
| | - Chao-Yang Zhang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People’s Hospital), Shanghai Jiao Tong University, Shanghai, China
- National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Diseases; Shanghai Engineering Center for Visual Science and Photomedicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Jing-Ting Zhang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People’s Hospital), Shanghai Jiao Tong University, Shanghai, China
- National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Diseases; Shanghai Engineering Center for Visual Science and Photomedicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Li-Min Gu
- Department of Ophthalmology, Shanghai Aier Eye Hospital, Shanghai, China
| | - Guo-Tong Xu
- Department of Ophthalmology of Tongji Hospital, Tongji Eye Institute, Department of Regenerative Medicine, and Department of Pharmacology, Tongji University School of Medicine, Shanghai, China
| | - Jing-Fa Zhang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People’s Hospital), Shanghai Jiao Tong University, Shanghai, China
- National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Diseases; Shanghai Engineering Center for Visual Science and Photomedicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| |
Collapse
|
28
|
Zhu X, Zhou C, Zhao S, Zheng Z. Role of m6A methylation in retinal diseases. Exp Eye Res 2023; 231:109489. [PMID: 37084873 DOI: 10.1016/j.exer.2023.109489] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 03/06/2023] [Accepted: 04/19/2023] [Indexed: 04/23/2023]
Abstract
Retinal diseases remain among the leading causes of visual impairment in developed countries, despite great efforts in prevention and early intervention. Due to the limited efficacy of current retinal therapies, novel therapeutic methods are urgently required. Over the past two decades, advances in next-generation sequencing technology have facilitated research on RNA modifications, which can elucidate the relevance of epigenetic mechanisms to disease. N6-methyladenosine (m6A), formed by methylation of adenosine at the N6-position, is the most widely studied RNA modification and plays an important role in RNA metabolism. It is dynamically regulated by writers (methyltransferases) and erasers (demethylases), and recognized by readers (m6A binding proteins). Although the discovery of m6A methylation can be traced back to the 1970s, its regulatory roles in retinal diseases are rarely appreciated. Here, we provide an overview of m6A methylation, and discuss its effects and possible mechanisms on retinal diseases, including diabetic retinopathy, age-related macular degeneration, retinoblastoma, retinitis pigmentosa, and proliferative vitreoretinopathy. Furthermore, we highlight potential agents targeting m6A methylation for retinal disease treatment and discuss the limitations and challenges of research in the field of m6A methylation.
Collapse
Affiliation(s)
- Xinyu Zhu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China; National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, 200080, China
| | - Chuandi Zhou
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China; National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, 200080, China
| | - Shuzhi Zhao
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China; National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, 200080, China.
| | - Zhi Zheng
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China; National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, 200080, China.
| |
Collapse
|
29
|
m6A Modification-Association with Oxidative Stress and Implications on Eye Diseases. Antioxidants (Basel) 2023; 12:antiox12020510. [PMID: 36830067 PMCID: PMC9952187 DOI: 10.3390/antiox12020510] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 02/09/2023] [Accepted: 02/15/2023] [Indexed: 02/19/2023] Open
Abstract
Oxidative stress (OS) refers to a state of imbalance between oxidation and antioxidation. OS is considered to be an important factor leading to aging and a range of diseases. The eyes are highly oxygen-consuming organs. Due to its continuous exposure to ultraviolet light, the eye is particularly vulnerable to the impact of OS, leading to eye diseases such as corneal disease, cataracts, glaucoma, etc. The N6-methyladenosine (m6A) modification is the most investigated RNA post-transcriptional modification and participates in a variety of cellular biological processes. In this study, we review the role of m6A modification in oxidative stress-induced eye diseases and some therapeutic methods to provide a relatively overall understanding of m6A modification in oxidative stress-related eye diseases.
Collapse
|
30
|
The Role of N6-Methyladenosine Modification in Microvascular Dysfunction. Cells 2022; 11:cells11203193. [PMID: 36291060 PMCID: PMC9600171 DOI: 10.3390/cells11203193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 09/24/2022] [Accepted: 09/28/2022] [Indexed: 11/16/2022] Open
Abstract
Microvascular dysfunction (MVD) has long plagued the medical field despite improvements in its prevention, diagnosis, and intervention. Microvascular lesions from MVD increase with age and further lead to impaired microcirculation, target organ dysfunction, and a mass of microvascular complications, thus contributing to a heavy medical burden and rising disability rates. An up-to-date understanding of molecular mechanisms underlying MVD will facilitate discoveries of more effective therapeutic strategies. Recent advances in epigenetics have revealed that RNA methylation, an epigenetic modification, has a pivotal role in vascular events. The N6-methylation of adenosine (m6A) modification is the most prevalent internal RNA modification in eukaryotic cells, which regulates vascular transcripts through splicing, degradation, translation, as well as translocation, thus maintaining microvascular homeostasis. Conversely, the disruption of the m6A regulatory network will lead to MVD. Herein, we provide a review discussing how m6A methylation interacts with MVD. We also focus on alterations of the m6A regulatory network under pathological conditions. Finally, we highlight the value of m6A regulators as prognostic biomarkers and novel therapeutic targets, which might be a promising addition to clinical medicine.
Collapse
|
31
|
Hu Q, Zhang X, Sun M, jiang B, Zhang Z, Sun D. Potential epigenetic molecular regulatory networks in ocular neovascularization. Front Genet 2022; 13:970224. [PMID: 36118885 PMCID: PMC9478661 DOI: 10.3389/fgene.2022.970224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 08/02/2022] [Indexed: 11/23/2022] Open
Abstract
Neovascularization is one of the many manifestations of ocular diseases, including corneal injury and vascular diseases of the retina and choroid. Although anti-VEGF drugs have been used to effectively treat neovascularization, long-term use of anti-angiogenic factors can cause a variety of neurological and developmental side effects. As a result, better drugs to treat ocular neovascularization are urgently required. There is mounting evidence that epigenetic regulation is important in ocular neovascularization. DNA methylation and histone modification, non-coding RNA, and mRNA modification are all examples of epigenetic mechanisms. In order to shed new light on epigenetic therapeutics in ocular neovascularization, this review focuses on recent advances in the epigenetic control of ocular neovascularization as well as discusses these new mechanisms.
Collapse
|
32
|
Huang C, Qi P, Cui H, Lu Q, Gao X. CircFAT1 regulates retinal pigment epithelial cell pyroptosis and autophagy via mediating m6A reader protein YTHDF2 expression in diabetic retinopathy. Exp Eye Res 2022; 222:109152. [PMID: 35714699 DOI: 10.1016/j.exer.2022.109152] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 05/28/2022] [Accepted: 06/10/2022] [Indexed: 11/04/2022]
Abstract
Diabetic retinopathy (DR) is a serious blinding complication of diabetes. At present, the therapeutic intervention effect is limited. We aimed to investigate the circRNA expression profiles in retinal proliferative fibrovascular membranes of patients with DR and explore the effect of circFAT1 on pyroptosis and autophagy of high glucose (HG)-induced retinal pigment epithelial (RPE) cells and its molecualr mechanism. In this study, circRNA sequencing was performed to determine the expression profiles of circRNAs in DR patients. The expression of circFAT1 was measured by qRT-PCR. Cell counting kit-8, transmission electron microscope, western blot, immunofluorescence and enzyme-linked immunosorbent assay were conducted to explore the roles of HG and circFAT1 in RPE cell pyroptosis and autophagy. RNA pull down was used to determine the binding protein of circFAT1. Our data showed that HG significantly reduced the viability of RPE cells, inhibited cell autophagy and contributed to cell pyroptosis. In addition, a total of 189 differentially expressed circRNAs (DEcircRNAs) were identified between DR patients and non-DR patients, including 93 upregulated and 96 downregulated DEcircRNAs in the retinal proliferative fibrovascular membranes of DR patients. Pathway analysis showed that DEcircRNAs were mainly involved in MAPK signaling pathway, TGF-beta signaling pathway and adherens junction. Moreover, circFAT1 was significantly downregulated in retinal proliferative fibrovascular membranes of DR patients and HG-induced RPE cells. CircFAT1 overexpression remarkably enhanced the expression of LC3B, while reduced the expression of GSDMD in HG-induced RPE cells. RNA pull down combined with western blot analysis indicated that circFAT1 bound to m6A reader YTHDF2. YTHDF2 overexpression significantly increased the protein expression of LC3B in HG-induced RPE cells. In summary, circFAT1 promoted autophagy and inhibited pyroptosis of RPE cells induced by HG, and could combine with YTHDF2. This study provides new ideas for DR prevention and treatment.
Collapse
Affiliation(s)
- Chengchi Huang
- Department of Ophthalmology, The Fourth Hospital of Harbin Medical University, 37 YiYuan Street, NanGang District, Harbin, 150001, China.
| | - Peng Qi
- Department of Ophthalmology, The Fourth Hospital of Harbin Medical University, 37 YiYuan Street, NanGang District, Harbin, 150001, China
| | - Hao Cui
- Department of Ophthalmology, Harbin 242 Hospital, 3 WeiJian Road, PingFang District, Harbin, China
| | - Qun Lu
- Department of Ophthalmology, Sino-Singapore Eco-city Hospital of TianJin Medical University, Tianjin, 300467, China
| | - Xue Gao
- Department of Ophthalmology, The Fourth Hospital of Harbin Medical University, 37 YiYuan Street, NanGang District, Harbin, 150001, China
| |
Collapse
|
33
|
Geng X, Li Z, Yang Y. Emerging Role of Epitranscriptomics in Diabetes Mellitus and Its Complications. Front Endocrinol (Lausanne) 2022; 13:907060. [PMID: 35692393 PMCID: PMC9184717 DOI: 10.3389/fendo.2022.907060] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 04/14/2022] [Indexed: 01/13/2023] Open
Abstract
Diabetes mellitus (DM) and its related complications are among the leading causes of disability and mortality worldwide. Substantial studies have explored epigenetic regulation that is involved in the modifications of DNA and proteins, but RNA modifications in diabetes are still poorly investigated. In recent years, posttranscriptional epigenetic modification of RNA (the so-called 'epitranscriptome') has emerged as an interesting field of research. Numerous modifications, mainly N6 -methyladenosine (m6A), have been identified in nearly all types of RNAs and have been demonstrated to have an indispensable effect in a variety of human diseases, such as cancer, obesity, and diabetes. Therefore, it is particularly important to understand the molecular basis of RNA modifications, which might provide a new perspective for the pathogenesis of diabetes mellitus and the discovery of new therapeutic targets. In this review, we aim to summarize the recent progress in the epitranscriptomics involved in diabetes and diabetes-related complications. We hope to provide some insights for enriching the understanding of the epitranscriptomic regulatory mechanisms of this disease as well as the development of novel therapeutic targets for future clinical benefit.
Collapse
Affiliation(s)
- Xinqian Geng
- Department of Endocrinology, The Affiliated Hospital of Yunnan University and the Second People’s Hospital of Yunnan Province, Kunming, China
| | - Zheng Li
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, China
| | - Ying Yang
- Department of Endocrinology, The Affiliated Hospital of Yunnan University and the Second People’s Hospital of Yunnan Province, Kunming, China
| |
Collapse
|
34
|
Chen Y, Xia Q, Zeng Y, Zhang Y, Zhang M. Regulations of Retinal Inflammation: Focusing on Müller Glia. Front Cell Dev Biol 2022; 10:898652. [PMID: 35573676 PMCID: PMC9091449 DOI: 10.3389/fcell.2022.898652] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 04/11/2022] [Indexed: 12/12/2022] Open
Abstract
Retinal inflammation underlies multiple prevalent retinal diseases. While microglia are one of the most studied cell types regarding retinal inflammation, growing evidence shows that Müller glia play critical roles in the regulation of retinal inflammation. Müller glia express various receptors for cytokines and release cytokines to regulate inflammation. Müller glia are part of the blood-retinal barrier and interact with microglia in the inflammatory responses. The unique metabolic features of Müller glia in the retina makes them vital for retinal homeostasis maintenance, regulating retinal inflammation by lipid metabolism, purine metabolism, iron metabolism, trophic factors, and antioxidants. miRNAs in Müller glia regulate inflammatory responses via different mechanisms and potentially regulate retinal regeneration. Novel therapies are explored targeting Müller glia for inflammatory retinal diseases treatment. Here we review new findings regarding the roles of Müller glia in retinal inflammation and discuss the related novel therapies for retinal diseases.
Collapse
Affiliation(s)
- Yingying Chen
- Department of Ophthalmology, Sichuan University West China Hospital, Sichuan University, Chengdu, China
- Research Laboratory of Macular Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Qinghong Xia
- Operating Room of Anesthesia Surgery Center, West China Hospital, Sichuan University, Chengdu, China
- West China School of Nursing, Sichuan University, Chengdu, China
| | - Yue Zeng
- Department of Ophthalmology, Sichuan University West China Hospital, Sichuan University, Chengdu, China
- Research Laboratory of Macular Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Yun Zhang
- Department of Ophthalmology, Sichuan University West China Hospital, Sichuan University, Chengdu, China
- Research Laboratory of Macular Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Meixia Zhang
- Department of Ophthalmology, Sichuan University West China Hospital, Sichuan University, Chengdu, China
- Research Laboratory of Macular Disease, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Meixia Zhang,
| |
Collapse
|
35
|
Chen T, Zhu W, Wang C, Dong X, Yu F, Su Y, Huang J, Huo L, Wan P. ALKBH5-Mediated m 6A Modification of A20 Regulates Microglia Polarization in Diabetic Retinopathy. Front Immunol 2022; 13:813979. [PMID: 35300330 PMCID: PMC8920977 DOI: 10.3389/fimmu.2022.813979] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 02/09/2022] [Indexed: 12/17/2022] Open
Abstract
Background To investigate the role of microglia polarization in the pathogenesis of diabetic retinopathy, and study the mechanism of ALKBH5-mediated m6A modification of A20 of retinal microglia polarization. Methods Diabetics rats were constructed and the M1/M2 polarization of retinal microglia was determined using immunofluorescence, flow cytometry, and quantitative real-time PCR (qRT-PCR). Glucose at different concentrations was added to treat the microglia, and the polarization rate was detected. RNA sequencing was performed to identify the differentially expressed gene in glucose treated microglia, and A20 expression was confirmed by qRT-PCR and western blotting. Lentiviruses encoding shRNA for A20 or overexpressing A20 were constructed to clarify the role of A20 in microglia polarization in vitro and vivo. N6-methyladenosine (m6A) modification level and degradation rate of A20 were determined and m6A related proteins were detected. Results Diabetics rats showed a higher M1 polarization rate but lower M2 polarization rate of retinal microglia. With the increase of glucose concentration, microglia tend to polarize into M1 inflammatory type rather than M2 anti-inflammatory type. Shown by RNA sequencing, glucose treated microglia showed a differentially expressed gene profile, which was enriched in kinds of inflammatory categories and pathways. A20 expression was lower in microglia with glucose treatment, which was demonstrated to negatively regulate the M1 polarization. Moreover, intraocular injection of A20-overexpression lentiviruses (OE-A20) rectified the enhanced M1 retinal microglia polarization of diabetes rats. The higher m6A modification level and faster degradation rate of A20 was observed in glucose treated microglia, which was mediated by m6A demethylase ALKBH5. Conclusion Lower expression A20 resulted in the enhanced M1 polarization of retinal microglia in diabetic retinopathy, which was caused by ALKBH5 mediated m6A modification. This study may provide new perspectives on not only the pathogenesis but also the diagnosis and treatment for diabetic retinopathy.
Collapse
Affiliation(s)
- Tingting Chen
- Department of Ophthalmology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wenhui Zhu
- Department of Ophthalmology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Congyao Wang
- Department of Ophthalmology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xia Dong
- Department of Ophthalmology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Fenfen Yu
- Department of Ophthalmology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yihua Su
- Department of Ophthalmology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jingwen Huang
- Department of Ophthalmology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lijun Huo
- Department of Ophthalmology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Pengxia Wan
- Department of Ophthalmology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
36
|
Wang Y, Xu M, Yue P, Zhang D, Tong J, Li Y. Novel Insights Into the Potential Mechanisms of N6-Methyladenosine RNA Modification on Sepsis-Induced Cardiovascular Dysfunction: An Update Summary on Direct and Indirect Evidences. Front Cell Dev Biol 2021; 9:772921. [PMID: 34869371 PMCID: PMC8633316 DOI: 10.3389/fcell.2021.772921] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 10/18/2021] [Indexed: 12/18/2022] Open
Abstract
Sepsis is a life-threatening organ dysfunction caused by a host’s dysfunctional response to infection. As is known to all, septic heart disease occurs because pathogens invading the blood stimulate the activation of endothelial cells, causing a large number of white blood cells to accumulate and trigger an immune response. However, in severe sepsis, the hematopoietic system is inhibited, and there will also be a decline in white blood cells, at which time the autoimmune system will also be suppressed. During the immune response, a large number of inflammatory factors are released into cells to participate in the inflammatory process, which ultimately damages cardiac myocytes and leads to impaired cardiac function. N6-methyladenosine (m6A) is a common RNA modification in mRNA and non-coding RNA that affects RNA splicing, translation, stability, and epigenetic effects of some non-coding RNAs. A large number of emerging evidences demonstrated m6A modification had been involved in multiple biological processes, especially for sepsis and immune disorders. Unfortunately, there are limited results provided to analyze the association between m6A modification and sepsis-induced cardiovascular dysfunction (SICD). In this review, we firstly summarized current evidences on how m6A mediates the pathophysiological process in cardiac development and cardiomyopathy to emphasize the importance of RNA methylation in maintaining heart biogenesis and homeostasis. Then, we clarified the participants of m6A modification in extended inflammatory responses and immune system activation, which are the dominant and initial changes secondary to sepsis attack. After that, we deeply analyzed the top causes of SICD and identified the activation of inflammatory cytokines, endothelial cell dysfunction, and mitochondrial failure. Thus, the highlight of this review is that we systematically collected all the related potential mechanisms between m6A modification and SICD causes. Although there is lack of direct evidences on SICD, indirect evidences had been demonstrated case by case on every particular molecular mechanism and signal transduction, which require further explorations into the potential links among the listed mechanisms. This provides novel insights into the understanding of SICD.
Collapse
Affiliation(s)
- Yang Wang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Miaomiao Xu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China.,Department of Immunology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Peng Yue
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Donghui Zhang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan, China
| | - Jiyu Tong
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China.,Department of Immunology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Yifei Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|