1
|
Liu YJ, Jia GR, Zhang SH, Guo YL, Ma XZ, Xu HM, Xie JX. The role of microglia in neurodegenerative diseases: from the perspective of ferroptosis. Acta Pharmacol Sin 2025:10.1038/s41401-025-01560-4. [PMID: 40307457 DOI: 10.1038/s41401-025-01560-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 04/06/2025] [Indexed: 05/02/2025]
Abstract
Iron plays a pivotal role in numerous fundamental biological processes in the brain. Among the various cell types in the central nervous system, microglia are recognized as the most proficient cells in accumulating and storing iron. Nonetheless, iron overload can induce inflammatory phenotype of microglia, leading to the production of proinflammatory cytokines and contributing to neurodegeneration. A growing body of evidence shows that disturbances in iron homeostasis in microglia is associated with a range of neurodegenerative disorders. Recent research has revealed that microglia are highly sensitive to ferroptosis, a form of iron-dependent cell death. How iron overload influences microglial function? Whether disbiosis in iron metabolism and ferroptosis in microglia are involved in neurodegenerative disorders and the underlying mechanisms remain to be elucidated. In this review we focus on the recent advances in research on microglial iron metabolism as well as ferroptosis in microglia. Meanwhile, we provide a comprehensive overview of the involvement of microglial ferroptosis in neurodegenerative disorders from the perspective of crosstalk between microglia and neuron, with a focus on Alzheimer's disease and Parkinson's disease.
Collapse
Affiliation(s)
- Ying-Juan Liu
- Institute of Brain Science and Disease, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, China
| | - Guo-Rui Jia
- Institute of Brain Science and Disease, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, China
- Department of Physiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Sheng-Han Zhang
- Institute of Brain Science and Disease, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, China
- Department of Physiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Yun-Liang Guo
- Institute of Brain Science and Disease, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, China
| | - Xi-Zhen Ma
- College of Life Sciences and Health, University of Health and Rehabilitation Science, Qingdao, 266113, China.
| | - Hua-Min Xu
- Institute of Brain Science and Disease, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, China.
- Department of Physiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China.
| | - Jun-Xia Xie
- Institute of Brain Science and Disease, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, China.
| |
Collapse
|
2
|
Liao Q, Li Y, Cui M, Liu M. m6A demethylase ALKBH5 reduces ferroptosis in diabetic retinopathy through the m6A-YTHDF1-ACSL4 axis. Diabet Med 2025:e70033. [PMID: 40210448 DOI: 10.1111/dme.70033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 03/18/2025] [Accepted: 03/22/2025] [Indexed: 04/12/2025]
Abstract
AIM Diabetic retinopathy (DR) represents the main ocular complication of diabetes. Targeting ferroptosis is a promising treatment of choice for various diabetic complications. N6-methyladenosine (m6A) demethylase alkylation repair homolog protein 5 (ALKBH5) functions as a pivotal regulator of ferroptosis, and we investigated its role and molecular mechanisms in ferroptosis in DR. METHODS A DR mouse model was developed by streptozotocin (STZ) intraperitoneal injection. High glucose (HG)-induced human retinal pigment epithelial cells (ARPE-19) were used as a DR model in vitro. ALKBH5, YTH N6-methyladenosine RNA binding protein 1 (YTHDF1) and acyl-CoA synthetase long-chain family member 4 (ACSL4) expression levels were examined by RT-qPCR and Western blot. The biological functions of ALKBH5 in vitro and in vivo were investigated by gain-of-function and loss-of-function analyses. ALKBH5's downstream regulatory mechanisms were detected by bioinformatics analysis, RNA pull-down, MeRIP-qPCR and actinomycin D assay. RESULTS ALKBH5 was under-expressed while YTHDF1 and ACSL4 were up-regulated in the retinal tissues of STZ-induced DR mice and HG-stimulated ARPE-19 cells. Ectopically expressed ALKBH5 or YTHDF1 knockdown partially reversed the increased ferroptosis in vitro and in vivo, evidenced by decreased levels of Fe2+, malondialdehyde and reactive oxygen species yet increased glutathione level. ALKBH5 mediated m6A modification of ACSL4 mRNA and disrupted its stability in a YTHDF1-dependent manner. Importantly, in vivo data demonstrated that overexpression of ALKBH5 or YTHDF1 knockdown repressed ferroptosis and alleviated DR by down-regulating ACSL4. CONCLUSION These findings suggest that ALKBH5 may delay DR progression by reducing ferroptosis through the m6A-YTHDF1-ACSL4 axis, offering therapeutic paradigms for the treatment of DR.
Collapse
Affiliation(s)
- Qinghui Liao
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong (Shenzhen) & Longgang District People's Hospital of Shenzhen, Shenzhen, China
| | - Yan Li
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong (Shenzhen) & Longgang District People's Hospital of Shenzhen, Shenzhen, China
| | - Meijing Cui
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong (Shenzhen) & Longgang District People's Hospital of Shenzhen, Shenzhen, China
| | - Min Liu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong (Shenzhen) & Longgang District People's Hospital of Shenzhen, Shenzhen, China
| |
Collapse
|
3
|
Lan J, Huang X, Li H, Lin S, Huang J, Yang W, Ouyang M, Fang J, Xu Q. YTHDF2 Regulates Advanced Glycation End Products-Induced Melanogenesis through Inhibiting A20 Expression in Human Dermal Fibroblasts. Inflammation 2025; 48:919-934. [PMID: 39009810 DOI: 10.1007/s10753-024-02097-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 05/06/2024] [Accepted: 07/01/2024] [Indexed: 07/17/2024]
Abstract
Fibroblast A20 suppresses advanced glycation end products (AGEs)-induced melanogenesis by inhibiting NLRP3 inflammasome activation. AGEs repress A20 expression and significantly m6A-methylate A20 mRNA in fibroblasts. YTHDF2 is the most studied m6A reader protein and can accelerate degradation of m6A-modified mRNA. Whether YTHDF2 regulates AGEs-induced A20 expression and pigmentation is unknown. In this study, we confirmed that YTHDF2 inversely regulated AGEs-BSA-inhibited A20 expression but facilitated AGEs-BSA-activated NF-κB signaling and NLRP3 inflammasome in fibroblasts via YTHDF2 knockdown and overexpression experiments. Mechanistically, YTHDF2 bound to m6A-modified A20 mRNA induced by AGEs-BSA and increased its degradation. Moreover, fibroblast YTHDF2 robustly promoted AGEs-BSA-induced IL-18 level in coculture supernatants and melanin content, tyrosinase activity, and expression of microphthalmia-associated transcription factor and tyrosinase in melanocytes, which were significantly blocked by IL-18 binding protein. Further, fibroblast YTHDF2 markedly increased AGEs-BSA-induced epidermal melanin level in cocultured ex vivo skin and MAPKs activation in melanocytes. Importantly, upregulated dermal YTHDF2 expression was negatively correlated with dermal A20 level and positively associated with both epidermal melanin and dermal AGEs content in sun-exposed skin and lesions of melasma and solar lentigo. These findings suggest that fibroblast YTHDF2 positively regulates AGEs-induced melanogenesis mainly via A20/ NF-κB /NLRP3 inflammasome/ IL-18 /MAPKs axis in an m6A-dependent manner and functions in photoaging-induced hyperpigmentation skin disorders.
Collapse
Affiliation(s)
- Jingjing Lan
- Department of Dermato-Venereology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, P. R. China
| | - Xianyin Huang
- Department of Dermato-Venereology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, P. R. China
| | - Hongpeng Li
- Department of Dermato-Venereology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, P. R. China
| | - Shen Lin
- Department of Dermato-Venereology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, P. R. China
| | - Jingqian Huang
- Department of Dermato-Venereology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, P. R. China
| | - Weixin Yang
- Department of Dermato-Venereology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, P. R. China
| | - Mengting Ouyang
- Department of Dermato-Venereology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, P. R. China
| | - Jiaqi Fang
- Department of Dermato-Venereology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, P. R. China.
| | - Qingfang Xu
- Department of Dermato-Venereology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, P. R. China.
| |
Collapse
|
4
|
Seo H, Park SJ, Song M. Diabetic Retinopathy (DR): Mechanisms, Current Therapies, and Emerging Strategies. Cells 2025; 14:376. [PMID: 40072104 PMCID: PMC11898816 DOI: 10.3390/cells14050376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 02/21/2025] [Accepted: 02/28/2025] [Indexed: 03/15/2025] Open
Abstract
Diabetic retinopathy (DR) is one of the most prevalent complications of diabetes, affecting nearly one-third of patients with diabetes mellitus and remaining a leading cause of blindness worldwide. Among the various diabetes-induced complications, DR is of particular importance due to its direct impact on vision and the irreversible damage to the retina. DR is characterized by multiple pathological processes, primarily a hyperglycemia-induced inflammatory response and oxidative stress. Current gold standard therapies, such as anti-VEGF injections and photocoagulation, have shown efficacy in slowing disease progression. However, challenges such as drug resistance, partial therapeutic responses, and the reliance on direct eye injections-which often result in low patient compliance-remain unresolved. This review provides a comprehensive overview of the underlying molecular mechanisms in DR, the current therapies, and their unmet needs for DR treatment. Additionally, emerging therapeutic strategies for improving DR treatment outcomes are discussed.
Collapse
Affiliation(s)
| | | | - Minsoo Song
- New Drug Development Center, Daegu-Gyeongbukk Medical Innovation Foundation (K-MEDI hub), 80 Cheombok-ro, Dong-gu, Daegu 41061, Republic of Korea; (H.S.); (S.-J.P.)
| |
Collapse
|
5
|
Han D, Liu J, Wang Y, Wang H, Yuan L, Jin W, Song L. Serum A20 level is associated with bone mineral density in male patients with type 2 diabetes mellitus. Front Endocrinol (Lausanne) 2025; 16:1490214. [PMID: 40078583 PMCID: PMC11899168 DOI: 10.3389/fendo.2025.1490214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 02/10/2025] [Indexed: 03/14/2025] Open
Abstract
Background A20, also known as TNF-α-induced protein 3 (TNFAIP3), is a crucial negative regulator of inflammation and immune responses. Emerging evidence suggests that A20 is involved in the regulation of glucose metabolism and plays a significant role in bone metabolic diseases by inhibiting nuclear factor (NF)-κB activation. However, the potential relationship between serum A20 level and bone mineral density (BMD) in patients with type 2 diabetes mellitus (T2DM) has not been explored. This study aims to investigate the association between serum A20 level with BMD and bone turnover markers (BTMs) in patients with T2DM. Method A total of 189 patients with T2DM and 183 non-diabetic individuals were included in the study based on the inclusion and exclusion criteria. Participants were categorized into normal BMD and low BMD groups. Baseline clinical histories were collected through face-to-face questionnaires. Participants underwent measurements of blood biochemistry and anthropometric, hand grip strength records and short physical performance battery (SPPB) assessment. Serum A20 level was quantified by enzyme-linked immunosorbent assay kit. Areal BMD was measured using dual-energy x-ray absorptiometry (DXA). A T-score of less than -1.0 at the lumbar spine 1-4, femoral neck and/or total hip was classified as low BMD. Results Serum A20 level was lower in patients with T2DM compared to controls [41.30 (29.91, 61.87) vs 76.01 (54.90, 109.64) pg/mL, P<0.001]. Bivariate correlation analysis revealed that A20 level was not associated with SPPB but negatively correlated with waist-to-hip ratio (WHR). Pearson correlation analysis showed A20 level was positively correlated with lumbar spine 1-4 BMD in male diabetic patients (r=0.253, P=0.032). Multivariate regression analysis showed a positive association between serum A20 level and lumbar spine 1-4 BMD (Beta=0.047; 95% CI: 0.007-0.086; P=0.024) after multivariate adjustment. Logistic regression analysis showed that lower serum A20 level predicted low BMD in male patients with T2DM (OR: 0.22; 95% CI: 0.09-0.59; P=0.002). Conclusions Type 2 diabetic patients exhibited lower serum A20 level compared to non-diabetic individuals. In male patients with T2DM, serum A20 level showed a significant positive correlation with lumbar spine 1-4 BMD and could serve as an independent negative predictor for low BMD.
Collapse
Affiliation(s)
- Dongxu Han
- Department of Endocrinology, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Institute of Osteoporosis and Metabolic Bone Diseases, School of Medicine, Tongji University, Shanghai, China
| | - Jingnan Liu
- Department of Endocrinology, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Institute of Osteoporosis and Metabolic Bone Diseases, School of Medicine, Tongji University, Shanghai, China
| | - Yu Wang
- Department of Endocrinology, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Institute of Osteoporosis and Metabolic Bone Diseases, School of Medicine, Tongji University, Shanghai, China
| | - Hongxia Wang
- Department of Endocrinology, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Institute of Osteoporosis and Metabolic Bone Diseases, School of Medicine, Tongji University, Shanghai, China
| | - Lingdan Yuan
- Department of Endocrinology, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Institute of Osteoporosis and Metabolic Bone Diseases, School of Medicine, Tongji University, Shanghai, China
| | - Wei Jin
- Department of Endocrinology, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Institute of Osteoporosis and Metabolic Bone Diseases, School of Medicine, Tongji University, Shanghai, China
| | - Lige Song
- Department of Endocrinology, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Institute of Osteoporosis and Metabolic Bone Diseases, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
6
|
Qiu M, Zhang W, Dai J, Sun W, Lai M, Tang S, Xu E, Ning Y, Zhan L. A20 negatively regulates necroptosis-induced microglia/macrophages polarization and mediates cerebral ischemic tolerance via inhibiting the ubiquitination of RIP3. Cell Death Dis 2024; 15:904. [PMID: 39695113 DOI: 10.1038/s41419-024-07293-2] [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: 05/23/2024] [Revised: 12/01/2024] [Accepted: 12/05/2024] [Indexed: 12/20/2024]
Abstract
Neuronal necroptosis appears to be suppressed by the deubiquitinating enzyme A20 and is capable to regulate the polarization of microglia/macrophages after cerebral ischemia. We have demonstrated that hypoxic preconditioning (HPC) can alleviate receptor interacting protein 3 (RIP3)-induced necroptosis in CA1 after transient global cerebral ischemia (tGCI). However, it is still unclear whether HPC serves to regulate the phenotypic polarization of microglia/macrophages after cerebral ischemia by mitigating neuronal necroptosis. We hence aim to elucidate the underlying mechanism(s) by which the ubiquitination of RIP3-dependent necroptosis regulated by A20 affects microglia/macrophages phenotype after cerebral ischemic tolerance. We found that microglia/macrophages in CA1 of rats underwent M1 and M2 phenotypic polarization in response to tGCI. Notably, the treatment with HPC, as well as inhibitors of necroptosis, including Nec-1 and mixed lineage kinase domain-like (MLKL) siRNA, attenuated neuroinflammation associated with M1 polarization of microglia/macrophages induced by tGCI. Mechanistically, HPC was revealed to upregulate A20 and in turn enhance the interaction between A20 and RIP3, thereby reducing K63-linked polyubiquitination of RIP3 in CA1 after tGCI. Consequently, RIP3-dependent necroptosis and the M1 polarization of microglia/macrophages were blocked either by HPC or via overexpression of A20 in neurons, which ultimately mitigated cerebral injury in CA1 after tGCI. These data support that A20 serves as a crucial mediator of microglia/macrophages polarization by suppressing neuronal necroptosis in a RIP3 ubiquitination-dependent manner after tGCI. Also, a novel mechanism by which HPC functions in cerebral ischemic tolerance is elucidated.
Collapse
Affiliation(s)
- Meiqian Qiu
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Wenhao Zhang
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jiahua Dai
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Weiwen Sun
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Meijing Lai
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Shiyi Tang
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - En Xu
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yuping Ning
- The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China.
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, China.
| | - Lixuan Zhan
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
7
|
Mao L, You J, Xie M, Hu Y, Zhou Q. Arginine Methylation of β-Catenin Induced by PRMT2 Aggravates LPS-Induced Cognitive Dysfunction and Depression-Like Behaviors by Promoting Ferroptosis. Mol Neurobiol 2024; 61:7796-7813. [PMID: 38430350 DOI: 10.1007/s12035-024-04019-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 02/04/2024] [Indexed: 03/03/2024]
Abstract
Depression is a prevalent and debilitating psychiatric disorder, imposing substantial societal and individual burdens. This study aims to investigate the involvement of ferroptosis and microglial polarization in the pathogenesis of depression, as well as the underlying mechanism. Increased protein arginine methyltransferase 2 (PRMT2) expression was observed in BV2 cells and the hippocampus following lipopolysaccharide (LPS) stimulation. Mechanistically, alkylation repair homolog protein 5 (ALKBH5)-mediated m6A modification enhanced the stability of PRMT2 mRNA. PRMT2 promoted arginine methylation of β-catenin and induced proteasomal degradation of β-catenin proteins, resulting in transcriptional inhibition of glutathione peroxidase 4 (GPX4). The upregulation of PRMT2 further accelerated microglia polarization by activating ferroptosis through the β-catenin-GPX4 axis. Depletion of PRMT2 improved LPS-induced depressive- and anxiety-like behaviors as well as cognitive impairment by inhibiting ferroptosis and M1 polarization of microglia. Our findings underscore the crucial involvement of the ALKBH5-PRMT2-β-catenin-GPX4 axis in ferroptosis and M1 polarization of microglia, thereby offering novel insights into the pathogenesis interventions for depression.
Collapse
Affiliation(s)
- Lei Mao
- Department of Anesthesiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, No. 32, West Second Section, First Ring Road, Chengdu, 610072, Sichuan, China
| | - Jiyue You
- Department of Anesthesiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, No. 32, West Second Section, First Ring Road, Chengdu, 610072, Sichuan, China
| | - Min Xie
- Department of Anesthesiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, No. 32, West Second Section, First Ring Road, Chengdu, 610072, Sichuan, China
| | - Yunxia Hu
- Department of Anesthesiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, No. 32, West Second Section, First Ring Road, Chengdu, 610072, Sichuan, China.
| | - Qin Zhou
- Department of Anesthesiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, No. 32, West Second Section, First Ring Road, Chengdu, 610072, Sichuan, China.
| |
Collapse
|
8
|
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
|
9
|
Liao Z, Wang J, Xu M, Li X, Xu H. The role of RNA m6A demethylase ALKBH5 in the mechanisms of fibrosis. Front Cell Dev Biol 2024; 12:1447135. [PMID: 39220683 PMCID: PMC11362088 DOI: 10.3389/fcell.2024.1447135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 08/06/2024] [Indexed: 09/04/2024] Open
Abstract
ALKBH5 is one of the demethylases involved in the regulation of RNA m6A modification. In addition to its role in the dynamic regulation of RNA m6A modification, ALKBH5 has been found to play important roles in various tissues fibrosis processes in recent years. However, the mechanisms and effects of ALKBH5 in fibrosis have been reported inconsistently. Multiple cell types, including parenchymal cells, immune cells (neutrophils and T cells), macrophages, endothelial cells, and fibroblasts, play roles in various stages of fibrosis. Therefore, this review analyzes the mechanisms by which ALKBH5 regulates these cells, its impact on their functions, and the outcomes of fibrosis. Furthermore, this review summarizes the role of ALKBH5 in fibrotic diseases such as pulmonary fibrosis, liver fibrosis, cardiac fibrosis, and renal fibrosis, and discusses various ALKBH5 inhibitors that have been discovered to date, exploring the potential of ALKBH5 as a clinical target for fibrosis.
Collapse
Affiliation(s)
| | | | | | - Xiaoyan Li
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Children’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongming Xu
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Children’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
10
|
Zhang Y, Zhou A. Macrophage activation contributes to diabetic retinopathy. J Mol Med (Berl) 2024; 102:585-597. [PMID: 38429382 DOI: 10.1007/s00109-024-02437-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 02/16/2024] [Accepted: 02/20/2024] [Indexed: 03/03/2024]
Abstract
Diabetic retinopathy (DR) is recognized as a neurovascular complication of diabetes, and emerging evidence underscores the pivotal role of inflammation in its pathophysiology. Macrophage activation is increasingly acknowledged as a key contributor to the onset and progression of DR. Different populations of macrophages originating from distinct sources contribute to DR-associated inflammation. Retinal macrophages can be broadly categorized into two main groups based on their origin: intrinsic macrophages situated within the retina and vitreoretinal interface and macrophages derived from infiltrating monocytes. The former comprises microglia (MG), perivascular macrophages, and macrophage-like hyalocytes. Retinal MG, as the principal population of tissue-resident population of mononuclear phagocytes, exhibits high heterogeneity and plasticity while serving as a crucial connector between retinal capillaries and synapses. This makes MG actively involved in the pathological processes across various stages of DR. Activated hyalocytes also contribute to the pathological progression of advanced DR. Additionally, recruited monocytes, displaying rapid turnover in circulation, augment the population of retinal macrophages during DR pathogenesis, exerting pathogenic or protective effect based on different subtypes. In this review, we examine novel perspectives on macrophage biology based on recent studies elucidating the diversity of macrophage identity and function, as well as the mechanisms influencing macrophage behavior. These insights may pave the way for innovative therapeutic strategies in the management of DR.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Ophthalmology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Aiyi Zhou
- Department of Ophthalmology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China.
| |
Collapse
|
11
|
Li H, Li B, Zheng Y. Role of microglia/macrophage polarisation in intraocular diseases (Review). Int J Mol Med 2024; 53:45. [PMID: 38551157 PMCID: PMC10998719 DOI: 10.3892/ijmm.2024.5369] [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: 11/18/2023] [Accepted: 03/05/2024] [Indexed: 04/02/2024] Open
Abstract
Macrophages form a crucial component of the innate immune system, and their activation is indispensable for various aspects of immune and inflammatory processes, tissue repair, and maintenance of the balance of the body's state. Macrophages are found in all ocular tissues, spanning from the front surface, including the cornea, to the posterior pole, represented by the choroid/sclera. The neural retina is also populated by specialised resident macrophages called microglia. The plasticity of microglia/macrophages allows them to adopt different activation states in response to changes in the tissue microenvironment. When exposed to various factors, microglia/macrophages polarise into distinct phenotypes, each exhibiting unique characteristics and roles. Furthermore, extensive research has indicated a close association between microglia/macrophage polarisation and the development and reversal of various intraocular diseases. The present article provides a review of the recent findings on the association between microglia/macrophage polarisation and ocular pathological processes (including autoimmune uveitis, optic neuritis, sympathetic ophthalmia, retinitis pigmentosa, glaucoma, proliferative vitreoretinopathy, subretinal fibrosis, uveal melanoma, ischaemic optic neuropathy, retinopathy of prematurity and choroidal neovascularization). The paradoxical role of microglia/macrophage polarisation in retinopathy of prematurity is also discussed. Several studies have shown that microglia/macrophages are involved in the pathology of ocular diseases. However, it is required to further explore the relevant mechanisms and regulatory processes. The relationship between the functional diversity displayed by microglia/macrophage polarisation and intraocular diseases may provide a new direction for the treatment of intraocular diseases.
Collapse
Affiliation(s)
- Haoran Li
- School of Opthalmology, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, P.R. China
| | - Biao Li
- School of Opthalmology, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, P.R. China
| | - Yanlin Zheng
- School of Opthalmology, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, P.R. China
| |
Collapse
|
12
|
Cai L, Xia M, Zhang F. Redox Regulation of Immunometabolism in Microglia Underpinning Diabetic Retinopathy. Antioxidants (Basel) 2024; 13:423. [PMID: 38671871 PMCID: PMC11047590 DOI: 10.3390/antiox13040423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/24/2024] [Accepted: 03/28/2024] [Indexed: 04/28/2024] Open
Abstract
Diabetic retinopathy (DR) is the leading cause of visual impairment and blindness among the working-age population. Microglia, resident immune cells in the retina, are recognized as crucial drivers in the DR process. Microglia activation is a tightly regulated immunometabolic process. In the early stages of DR, the M1 phenotype commonly shifts from oxidative phosphorylation to aerobic glycolysis for energy production. Emerging evidence suggests that microglia in DR not only engage specific metabolic pathways but also rearrange their oxidation-reduction (redox) system. This redox adaptation supports metabolic reprogramming and offers potential therapeutic strategies using antioxidants. Here, we provide an overview of recent insights into the involvement of reactive oxygen species and the distinct roles played by key cellular antioxidant pathways, including the NADPH oxidase 2 system, which promotes glycolysis via enhanced glucose transporter 4 translocation to the cell membrane through the AKT/mTOR pathway, as well as the involvement of the thioredoxin and nuclear factor E2-related factor 2 antioxidant systems, which maintain microglia in an anti-inflammatory state. Therefore, we highlight the potential for targeting the modulation of microglial redox metabolism to offer new concepts for DR treatment.
Collapse
Affiliation(s)
- Luwei Cai
- National Clinical Research Center for Eye Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China; (L.C.); (M.X.)
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai 200080, China
- Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai 200080, China
- Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai 200080, China
| | - Mengxue Xia
- National Clinical Research Center for Eye Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China; (L.C.); (M.X.)
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai 200080, China
- Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai 200080, China
- Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai 200080, China
| | - Fang Zhang
- National Clinical Research Center for Eye Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China; (L.C.); (M.X.)
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai 200080, China
- Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai 200080, China
- Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai 200080, China
| |
Collapse
|
13
|
Guo X, Qiu W, Li B, Qi Y, Wang S, Zhao R, Cheng B, Han X, Du H, Pan Z, Zhao S, Qiu J, Li G, Xue H. Hypoxia-Induced Neuronal Activity in Glioma Patients Polarizes Microglia by Potentiating RNA m6A Demethylation. Clin Cancer Res 2024; 30:1160-1174. [PMID: 37855702 DOI: 10.1158/1078-0432.ccr-23-0430] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 08/22/2023] [Accepted: 10/17/2023] [Indexed: 10/20/2023]
Abstract
PURPOSE Neuronal activity in the brain has been reported to promote the malignant progression of glioma cells via nonsynaptic paracrine and electrical synaptic integration mechanisms. However, the interaction between neuronal activity and the immune microenvironment in glioblastoma (GBM) remains largely unclear. EXPERIMENTAL DESIGN By applying chemogenetic techniques, we enhanced and inhibited neuronal activity in vitro and in a mouse model to study how neuronal activity regulates microglial polarization and affects GBM progression. RESULTS We demonstrate that hypoxia drove glioma stem cells (GSC) to produce higher levels of glutamate, which activated local neurons. Neuronal activity promoted GBM progression by facilitating microglial M2 polarization through enriching miR-200c-3p in neuron-derived exosomes, which decreased the expression of the m6A writer zinc finger CCCH-type containing 13 (ZC3H13) in microglia, impairing methylation of dual specificity phosphatase 9 (DUSP9) mRNA. Downregulation of DUSP9 promoted ERK pathway activation, which subsequently induced microglial M2 polarization. In the mouse model, cortical neuronal activation promoted microglial M2 polarization whereas cortical neuronal inhibition decreased microglial M2 polarization in GBM xenografts. miR-200c-3p knockdown in cortical neurons impaired microglial M2 polarization and GBM xenograft growth, even when cortical neurons were activated. Treatment with the anti-seizure medication levetiracetam impaired neuronal activation and subsequently reduced neuron-mediated microglial M2 polarization. CONCLUSIONS These findings indicated that hypoxic GSC-induced neuron activation promotes GBM progression by polarizing microglia via the exosomal miR-200c-3p/ZC3H13/DUSP9/p-ERK pathway. Levetiracetam, an antiepileptic drug, blocks the abnormal activation of neurons in GBM and impairs activity-dependent GBM progression. See related commentary by Cui et al., p. 1073.
Collapse
Affiliation(s)
- Xiaofan Guo
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong, China
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, Shandong, China
- Department of Neurology, Loma Linda University Health, Loma Linda, California
| | - Wei Qiu
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong, China
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, Shandong, China
| | - Boyan Li
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong, China
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, Shandong, China
| | - Yanhua Qi
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong, China
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, Shandong, China
| | - Shaobo Wang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong, China
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, Shandong, China
| | - Rongrong Zhao
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong, China
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, Shandong, China
| | - Bo Cheng
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, Shandong, China
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Xiao Han
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong, China
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, Shandong, China
- Department of Neurosurgery, Jinan Children's Hospital, Jinan, Shandong, China
| | - Hao Du
- Department of Cell Biology, University of Connecticut School of Medicine, Farmington, Connecticut
| | - Ziwen Pan
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong, China
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, Shandong, China
| | - Shulin Zhao
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong, China
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, Shandong, China
| | - Jiawei Qiu
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong, China
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, Shandong, China
| | - Gang Li
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong, China
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, Shandong, China
| | - Hao Xue
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong, China
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, Shandong, China
| |
Collapse
|
14
|
Huang Z, Zhang Y, Wang S, Qi R, Tao Y, Sun Y, Jiang D, Jiang X, Tao J. FOXD3-mediated transactivation of ALKBH5 promotes neuropathic pain via m 6A-dependent stabilization of 5-HT3A mRNA in sensory neurons. Proc Natl Acad Sci U S A 2024; 121:e2312861121. [PMID: 38285939 PMCID: PMC10861880 DOI: 10.1073/pnas.2312861121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 12/11/2023] [Indexed: 01/31/2024] Open
Abstract
The N6-methyladenosine (m6A) modification of RNA is an emerging epigenetic regulatory mechanism that has been shown to participate in various pathophysiological processes. However, its involvement in modulating neuropathic pain is still poorly understood. In this study, we elucidate a functional role of the m6A demethylase alkylation repair homolog 5 (ALKBH5) in modulating trigeminal-mediated neuropathic pain. Peripheral nerve injury selectively upregulated the expression level of ALKBH5 in the injured trigeminal ganglion (TG) of rats. Blocking this upregulation in injured TGs alleviated trigeminal neuropathic pain, while mimicking the upregulation of ALKBH5 in intact TG neurons sufficiently induced pain-related behaviors. Mechanistically, histone deacetylase 11 downregulation induced by nerve injury increases histone H3 lysine 27 acetylation (H3K27ac), facilitating the binding of the transcription factor forkhead box protein D3 (FOXD3) to the Alkbh5 promoter and promoting Alkbh5 transcription. The increased ALKBH5 erases m6A sites in Htr3a messenger RNA (mRNA), resulting in an inability of YT521-B homology domain 2 (YTHDF2) to bind to Htr3a mRNA, thus causing an increase in 5-HT3A protein expression and 5-HT3 channel currents. Conversely, blocking the increased expression of ALKBH5 in the injured TG destabilizes nerve injury-induced 5-HT3A upregulation and reverses mechanical allodynia, and the effect can be blocked by 5-HT3A knockdown. Together, FOXD3-mediated transactivation of ALKBH5 promotes neuropathic pain through m6A-dependent stabilization of Htr3a mRNA in TG neurons. This mechanistic understanding may advance the discovery of new therapeutic targets for neuropathic pain management.
Collapse
Affiliation(s)
- Zitong Huang
- Department of Physiology and Neurobiology, Suzhou Medical College of Soochow University, Suzhou215123, People’s Republic of China
- Centre for Ion Channelopathy, Soochow University, Suzhou215123, People’s Republic of China
| | - Yuan Zhang
- Clinical Research Center of Neurological Disease, Department of Geriatrics, The Second Affiliated Hospital of Soochow University, Suzhou215004, People’s Republic of China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou215123, People’s Republic of China
| | - Shoupeng Wang
- Department of Physiology and Neurobiology, Suzhou Medical College of Soochow University, Suzhou215123, People’s Republic of China
- Centre for Ion Channelopathy, Soochow University, Suzhou215123, People’s Republic of China
| | - Renfei Qi
- Department of Physiology and Neurobiology, Suzhou Medical College of Soochow University, Suzhou215123, People’s Republic of China
- Centre for Ion Channelopathy, Soochow University, Suzhou215123, People’s Republic of China
| | - Yu Tao
- Department of Physiology and Neurobiology, Suzhou Medical College of Soochow University, Suzhou215123, People’s Republic of China
- Centre for Ion Channelopathy, Soochow University, Suzhou215123, People’s Republic of China
| | - Yufang Sun
- Department of Physiology and Neurobiology, Suzhou Medical College of Soochow University, Suzhou215123, People’s Republic of China
- Centre for Ion Channelopathy, Soochow University, Suzhou215123, People’s Republic of China
| | - Dongsheng Jiang
- Institute of Regenerative Biology and Medicine, Helmholtz Zentrum München, Munich81377, Germany
| | - Xinghong Jiang
- Department of Physiology and Neurobiology, Suzhou Medical College of Soochow University, Suzhou215123, People’s Republic of China
- Centre for Ion Channelopathy, Soochow University, Suzhou215123, People’s Republic of China
| | - Jin Tao
- Department of Physiology and Neurobiology, Suzhou Medical College of Soochow University, Suzhou215123, People’s Republic of China
- Centre for Ion Channelopathy, Soochow University, Suzhou215123, People’s Republic of China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou215123, People’s Republic of China
- Ministry of Education (MOE) Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou215123, People’s Republic of China
| |
Collapse
|
15
|
Wang D, Qian W, Wu D, Wu Y, Lu K, Zou G. METTL3 promotes microglial inflammation via MEF2C in spinal cord injury. Cell Tissue Res 2024; 395:189-197. [PMID: 38180567 DOI: 10.1007/s00441-023-03855-6] [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: 03/31/2023] [Accepted: 12/20/2023] [Indexed: 01/06/2024]
Abstract
Spinal cord injury (SCI) is a significant contributor to disability in contemporary society, resulting in substantial psychological and economic burdens for patients and their family. Microglia-mediated inflammation is an important factor affecting the nerve repair of SCI patients. N6-methyladenosine (m6A) is a prevalent epigenetic modification in mammals, which shows a strong association with inflammation. However, the mechanism of m6A modification regulating microglia-mediated inflammation is still unclear. Here, we observed that METTL3, a m6A methylase, was increased in SCI mice and lipopolysaccharide (LPS)-exposed BV2 cells. Knockdown of METTL3 inhibited the increased expression of iNOS and IL-1β induced by LPS in vitro. Subsequently, MEF2C, myocyte-specific enhancer factor 2C, was decreased in SCI mice and LPS-exposed BV2 cells. Knockdown of MEF2C promoted the expression of iNOS and IL-1β. Sequence analysis showed that there were multiple highly confident m6A modification sites on the MEF2C mRNA. METTL3 antibody could pull down a higher level of MEF2C mRNA than the IgG in RNA binding protein immunoprecipitation assay. Knockdown of METTL3 promoted MEF2C protein expression and MEF2C mRNA expression, accompanied by a reduced m6A modification level on the MEF2C mRNA. Knockdown of MEF2C inhibited the anti-inflammatory effect of METTL3 siRNA. Our results suggest that METTL3 promotes microglia inflammation via regulating MEF2C mRNA m6A modification induced by SCI and LPS treatment.
Collapse
Affiliation(s)
- Dongliang Wang
- Department of Spinal Surgery, Yancheng First People's Hospital, Yancheng, 224006, China
| | - Wei Qian
- Department of Infectious Diseases, Yancheng Third People's Hospital, Yancheng, 224051, China
| | - Duanrong Wu
- Department of Orthopedics, Yancheng First People's Hospital, Yancheng, 224006, China
| | - Ya Wu
- Department of Orthopedics, Yancheng First People's Hospital, Yancheng, 224006, China
| | - Kun Lu
- Department of Orthopedics, Yancheng First People's Hospital, Yancheng, 224006, China
| | - Guoyou Zou
- Department of Orthopedics, Yancheng First People's Hospital, Yancheng, 224006, China.
| |
Collapse
|
16
|
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
|
17
|
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
|
18
|
Teng Y, Yi J, Chen J, Yang L. N6-Methyladenosine (m6A) Modification in Natural Immune Cell-Mediated Inflammatory Diseases. J Innate Immun 2023; 15:804-821. [PMID: 37903470 PMCID: PMC10673353 DOI: 10.1159/000534162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 09/13/2023] [Indexed: 11/01/2023] Open
Abstract
The post-transcriptional N6-methyladenosine (m6A) modification of RNA influences stability, transport, and translation with implications for various physiological and pathological processes. Immune cell development, differentiation, and activation are also thought to be regulated by m6A and affect host defense against pathogens and inflammatory response with impacts on infectious, neoplastic, autoimmune, cardiovascular, hepatic, and osteal diseases. The current review summarizes recent research on m6A in monocyte/macrophages, neutrophils, dendritic cells, natural killer cells, and microglia and gives insights into epigenetic modifications of the immune system and novel therapeutic strategies for immune-related diseases.
Collapse
Affiliation(s)
- Yan Teng
- Institute of Neurology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Jin Yi
- Center for Informational Biology, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Junnian Chen
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Lu Yang
- Institute of Neurology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
19
|
Yao Y, Li J, Zhou Y, Wang S, Zhang Z, Jiang Q, Li K. Macrophage/microglia polarization for the treatment of diabetic retinopathy. Front Endocrinol (Lausanne) 2023; 14:1276225. [PMID: 37842315 PMCID: PMC10569308 DOI: 10.3389/fendo.2023.1276225] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 09/07/2023] [Indexed: 10/17/2023] Open
Abstract
Macrophages/microglia are immune system defense and homeostatic cells that develop from bone marrow progenitor cells. According to the different phenotypes and immune responses of macrophages (Th1 and Th2), the two primary categories of polarized macrophages/microglia are those conventionally activated (M1) and alternatively activated (M2). Macrophage/microglial polarization is a key regulating factor in the development of inflammatory disorders, cancers, metabolic disturbances, and neural degeneration. Macrophage/microglial polarization is involved in inflammation, oxidative stress, pathological angiogenesis, and tissue healing processes in ocular diseases, particularly in diabetic retinopathy (DR). The functional phenotypes of macrophages/microglia affect disease progression and prognosis, and thus regulate the polarization or functional phenotype of microglia at different DR stages, which may offer new concepts for individualized therapy of DR. This review summarizes the involvement of macrophage/microglia polarization in physiological situations and in the pathological process of DR, and discusses the promising role of polarization in personalized treatment of DR.
Collapse
Affiliation(s)
- Yujia Yao
- Department of Ophthalmology, The Affiliated Eye Hospital of Nanjing Medical University, Nanjing, China
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Jiajun Li
- Department of Ophthalmology, The Affiliated Eye Hospital of Nanjing Medical University, Nanjing, China
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Yunfan Zhou
- Department of Ophthalmology, The Affiliated Eye Hospital of Nanjing Medical University, Nanjing, China
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Suyu Wang
- Department of Ophthalmology, The Affiliated Eye Hospital of Nanjing Medical University, Nanjing, China
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Ziran Zhang
- Department of Ophthalmology, The Affiliated Eye Hospital of Nanjing Medical University, Nanjing, China
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Qin Jiang
- Department of Ophthalmology, The Affiliated Eye Hospital of Nanjing Medical University, Nanjing, China
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Keran Li
- Department of Ophthalmology, The Affiliated Eye Hospital of Nanjing Medical University, Nanjing, China
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, China
| |
Collapse
|
20
|
Yu F, Wang C, Su Y, Chen T, Zhu W, Dong X, Ke W, Cai L, Yang S, Wan P. Comprehensive analysis of ferritinophagy-related genes and immune infiltration landscape in diabetic retinopathy. Front Endocrinol (Lausanne) 2023; 14:1177488. [PMID: 37522124 PMCID: PMC10377661 DOI: 10.3389/fendo.2023.1177488] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 06/28/2023] [Indexed: 08/01/2023] Open
Abstract
Background Diabetic retinopathy (DR) is deemed a microangiopathy and neurodegenerative disorder, which is a primary reason of visual impairment in the world. Ferritinophagy is a critical regulator of ferroptosis and has a vital part in the etiopathogenesis of DR. Nevertheless, its molecular mechanism in DR remains to be expounded. Methods The GSE146615 dataset was adopted to identify ferritinophagy-related differentially expressed genes (FRDEGs). The interactions and biological functions of the genes were described by means of functional enrichment analysis (FEA). The enriched gene sets were analyzed utilizing gene set enrichment analysis (GSEA) and gene set variation analysis (GSVA). Identification of hub genes was performed utilizing protein-protein interaction (PPI) analysis. mRNA-miRNA, mRNA-transcription factors (TF), mRNA-drugs, mRNA-RNA-binding proteins (RBP) interaction networks were constructed. In addition, datasets GSE60436 and GSE94019 were utilized for validation. The diagnostic performance of FRDEGs was assessed by means of receiver-operating characteristic curve monofactor analysis, followed by immune infiltration analysis. Lastly, quantitative real-time polymerase chain reaction (qRT-PCR) was implemented to analyze the validation of genes. Results In total, the identification of eight FRDEGs was completed utilizing differential expression analysis. FEA mainly implicated the autophagy of mitochondrion, mitochondrion disassembly, autophagosome assembly, and organization pathways. GSEA and GSVA mainly implicated the interferon alpha response, ultraviolet response up, interferon gamma response, apical junction, pical surface, and allograft rejection pathways. BECN1 and HERC2 displayed high diagnostic accuracies in validation sets. Immune infiltration analysis revealed that several immune cells related to ferritinophagy may be play potential roles in DR. Finally, qRT-PCR was utilized to validate the upregulated expression of BECN1 as well as the downregulated expression of BCAT2 and ATG7 in the DR model. Conclusion BECN1, HERC2, ATG7, and BCAT2 act as potential biomarkers for DR and might regulate ferritinophagy and the immune microenvironment to influence its development and progression. This research can provide new insights into pathogenesis of DR related to ferritinophagy.
Collapse
Affiliation(s)
- Fenfen Yu
- 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
| | - Yihua Su
- Department of Ophthalmology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - 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
| | - Xia Dong
- Department of Ophthalmology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wanyi Ke
- Department of Ophthalmology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Leqi Cai
- Department of Ophthalmology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shasha Yang
- Department of Ophthalmology, Guangzhou First People’s Hospital, The Second Affiliated Hospital, South China University of Technology, Guangzhou, China
| | - Pengxia Wan
- Department of Ophthalmology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
21
|
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
|
22
|
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
|
23
|
Yang X, Huang Z, Xu M, Chen Y, Cao M, Yi G, Fu M. Autophagy in the retinal neurovascular unit: New perspectives into diabetic retinopathy. J Diabetes 2023; 15:382-396. [PMID: 36864557 PMCID: PMC10172025 DOI: 10.1111/1753-0407.13373] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 02/08/2023] [Accepted: 02/18/2023] [Indexed: 03/04/2023] Open
Abstract
Diabetic retinopathy (DR) is one of the most prevalent retinal disorders worldwide, and it is a major cause of vision impairment in individuals of productive age. Research has demonstrated the significance of autophagy in DR, which is a critical intracellular homeostasis mechanism required for the destruction and recovery of cytoplasmic components. Autophagy maintains the physiological function of senescent and impaired organelles under stress situations, thereby regulating cell fate via various signals. As the retina's functional and fundamental unit, the retinal neurovascular unit (NVU) is critical in keeping the retinal environment's stability and supporting the needs of retinal metabolism. However, autophagy is essential for the normal NVU structure and function. We discuss the strong association between DR and autophagy in this review, as well as the many kinds of autophagy and its crucial physiological activities in the retina. By evaluating the pathological changes of retinal NVU in DR and the latest advancements in the molecular mechanisms of autophagy that may be involved in the pathophysiology of DR in NVU, we seek to propose new ideas and methods for the prevention and treatment of DR.
Collapse
Affiliation(s)
- Xiongyi Yang
- Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, P. R. China
- The Second Clinical School, Southern Medical University, Guangzhou, Guangdong, P. R. China
| | - Zexin Huang
- Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, P. R. China
- The Second Clinical School, Southern Medical University, Guangzhou, Guangdong, P. R. China
| | - Mei Xu
- The Second People's Hospital of Jingmen, Jingmen, Hubei, People's Republic of China
| | - Yanxia Chen
- Department of Ophthalmology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, P. R. China
| | - Mingzhe Cao
- Department of Ophthalmology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, P. R. China
| | - Guoguo Yi
- Department of Ophthalmology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, P. R. China
| | - Min Fu
- Department of Ophthalmology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, P. R. China
| |
Collapse
|
24
|
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
|
25
|
Liu D, Fan B, Li J, Sun T, Ma J, Zhou X, Feng S. N6-methyladenosine modification: A potential regulatory mechanism in spinal cord injury. Front Cell Neurosci 2022; 16:989637. [PMID: 36212687 PMCID: PMC9539101 DOI: 10.3389/fncel.2022.989637] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 09/05/2022] [Indexed: 12/03/2022] Open
Abstract
N6-methyladenosine (m6A), an essential post-transcriptional modification in eukaryotes, is closely related to the development of pathological processes in neurological diseases. Notably, spinal cord injury (SCI) is a serious traumatic disease of the central nervous system, with a complex pathological mechanism which is still not completely understood. Recent studies have found that m6A modification levels are changed after SCI, and m6A-related regulators are involved in the changes of the local spinal cord microenvironment after injury. However, research on the role of m6A modification in SCI is still in the early stages. This review discusses the latest progress in the dynamic regulation of m6A modification, including methyltransferases (“writers”), demethylases (“erasers”) and m6A -binding proteins (“readers”). And then analyses the pathological mechanism relationship between m6A and the microenvironment after SCI. The biological processes involved included cell death, axon regeneration, and scar formation, which provides new insight for future research on the role of m6A modification in SCI and the clinical transformation of strategies for promoting recovery of spinal cord function.
Collapse
Affiliation(s)
- Derong Liu
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, Tianjin, China
| | - Baoyou Fan
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, Tianjin, China
| | - Jinze Li
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, Tianjin, China
| | - Tao Sun
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, Tianjin, China
| | - Jun Ma
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, Tianjin, China
| | - Xianhu Zhou
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo, China
- *Correspondence: Xianhu Zhou,
| | - Shiqing Feng
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, Tianjin, China
- Shiqing Feng,
| |
Collapse
|
26
|
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
|