1
|
Zheng Y, Gu H, Kong Y. Targeting PTEN in ischemic stroke: From molecular mechanisms to therapeutic potentials. Exp Neurol 2024; 383:115023. [PMID: 39461709 DOI: 10.1016/j.expneurol.2024.115023] [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: 07/09/2024] [Revised: 10/20/2024] [Accepted: 10/20/2024] [Indexed: 10/29/2024]
Abstract
Ischemic stroke remains a leading cause of mortality and disability worldwide, driven by complex pathophysiological mechanisms, including excitotoxicity, oxidative stress, apoptosis, and neuroinflammation. PTEN (Phosphatase and tensin homolog deleted on chromosome 10) plays a crucial role in these processes, influencing key signaling pathways such as PI3K/Akt and mTOR. This review aims to explore PTEN's multifaceted functions in ischemic stroke, examining its interactions with non-coding RNAs, involvement in mitophagy and immune suppression, and overall impact on cellular homeostasis. We will investigate various therapeutic strategies targeting PTEN, including synthetic drugs, natural products, and exosome-based therapies enriched with specific miRNAs. Additionally, we will assess the potential of non-pharmaceutical interventions such as electroacupuncture, exercise, transcranial direct current stimulation (tDCS), and therapeutic hypothermia in modulating PTEN activity to enhance cererbroprotection and functional recovery. By elucidating these aspects, this review aims to inspire and motivate the audience in their research and clinical practice, highlighting PTEN as a promising therapeutic target and paving the way for developing effective treatments for ischemic stroke.
Collapse
Affiliation(s)
- Yane Zheng
- Department of Neurology, Shanghai Jiangong Hospital, Shanghai 200083, China.
| | - Huiying Gu
- Department of Internal Medicine, Tangqiao Community Health Service Center, Shanghai 200127, China
| | - Yuming Kong
- Department of Neurology, Yangpu Hospital, Tongji University School of Medicine, Shanghai 200438, China
| |
Collapse
|
2
|
Chen Y, Long T, Chen J, Wei H, Meng J, Kang M, Wang J, Zhang X, Xu Q, Zhang C, Xiong K. WTAP participates in neuronal damage by protein translation of NLRP3 in an m6A-YTHDF1-dependent manner after traumatic brain injury. Int J Surg 2024; 110:5396-5408. [PMID: 38874470 PMCID: PMC11392096 DOI: 10.1097/js9.0000000000001794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Accepted: 05/30/2024] [Indexed: 06/15/2024]
Abstract
BACKGROUND Traumatic brain injury (TBI) is a common complication of acute and severe neurosurgery. Remodeling of N6-methyladenosine (m6A) stabilization may be an attractive treatment option for neurological dysfunction after TBI. In the present study, the authors explored the epigenetic methylation of RNA-mediated NLRP3 inflammasome activation after TBI. METHODS Neurological dysfunction, histopathology, and associated molecules were examined in conditional knockout (CKO) WTAP [flox/flox, Camk2a-cre] , WTAP flox/flox , and pAAV-U6-shRNA-YTHDF1-transfected mice. Primary neurons were used in vitro to further explore the molecular mechanisms of action of WTAP/YTHDF1 following neural damage. RESULTS The authors found that WTAP and m6A levels were upregulated at an early stage after TBI, and conditional deletion of WTAP in neurons did not affect neurological function but promoted functional recovery after TBI. Conditional deletion of WTAP in neurons suppressed neuroinflammation at the TBI early phase: WTAP could directly act on NLRP3 mRNA, regulate NLRP3 mRNA m6A level, and promote NLRP3 expression after neuronal injury. Further investigation found that YTH domain of YTHDF1 could directly bind to NLRP3 mRNA and regulate NLRP3 protein expression. YTHDF1 mutation or silencing improved neuronal injury, inhibited Caspase-1 activation, and decreased IL-1β levels. This effect was mediated via suppression of NLRP3 protein translation, which also reversed the stimulative effect of WTAP overexpression on NLRP3 expression and inflammation. CONCLUSIONS Our results indicate that WTAP participates in neuronal damage by protein translation of NLRP3 in an m6A-YTHDF1-dependent manner after TBI and that WTAP/m6A/YTHDF1 downregulation therapeutics is a viable and promising approach for preserving neuronal function after TBI, which can provide support for targeted drug development.
Collapse
Affiliation(s)
- Yuhua Chen
- Department of Neurosurgery, Bijie Traditional Chinese Medical Hospital, Bijie, Guizhou
- Department of Central Laboratory, Xi'an Peihua University, Xi'an, Shaanxi
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan
| | - Tianlin Long
- Department of Neurosurgery, Bijie Traditional Chinese Medical Hospital, Bijie, Guizhou
| | - Junhui Chen
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan
- Department of Neurosurgery, Wuxi Clinical College of Anhui Medical University, 904 Hospital of Joint Logistic Support Force of PLA, Wuxi, Jiangsu Province
| | - Hong Wei
- Department of Rehabilitation Teaching and Research, Xi'an Siyuan University, Xi'an
| | - Jiao Meng
- Department of Neurosurgery, Bijie Traditional Chinese Medical Hospital, Bijie, Guizhou
- Department of Central Laboratory, Xi'an Peihua University, Xi'an, Shaanxi
| | - Meili Kang
- Department of Central Laboratory, Xi'an Peihua University, Xi'an, Shaanxi
| | - Juning Wang
- Department of Central Laboratory, Xi'an Peihua University, Xi'an, Shaanxi
| | - Xin Zhang
- Department of Neurosurgery, Bijie Traditional Chinese Medical Hospital, Bijie, Guizhou
| | - Quanhua Xu
- Department of Neurosurgery, Bijie Traditional Chinese Medical Hospital, Bijie, Guizhou
| | - Chi Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha
| | - Kun Xiong
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan
- Key Laboratory of Emergency and Trauma, Ministry of Education, College of Emergency and Trauma, Hainan Medical University, Haikou, Hainan
- Hunan Key Laboratory of Ophthalmology, Changsha, Hunan, People's Republic of China
| |
Collapse
|
3
|
Fan J, Zhong L, Yan F, Li X, Li L, Zhao H, Han Z, Wang R, Tao Z, Zheng Y, Ma Q, Luo Y. Alteration of N6-methyladenosine modification profiles in the neutrophilic RNAs following ischemic stroke. Neuroscience 2024; 553:56-73. [PMID: 38945353 DOI: 10.1016/j.neuroscience.2024.06.014] [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: 03/20/2024] [Revised: 06/04/2024] [Accepted: 06/18/2024] [Indexed: 07/02/2024]
Abstract
BACKGROUND N6-methyladenosine (m6A) is one of the most extensive RNA methylation modifications in eukaryotes and participates in the pathogenesis of numerous diseases including ischemic stroke. Peripheral blood neutrophils are forerunners after ischemic brain injury and exert crucial functions. This study aims to explore the transcriptional profiles of m6A modification in neutrophils of patients with ischemic stroke. RESULTS We found that the expression levels of m6A regulators FTO and YTHDC1 were notably decreased in the neutrophils following ischemic stroke, and FTO expression was negatively correlated with neutrophil counts and neutrophil-to-lymphocyte ratio (NLR). The m6A mRNA&lncRNA epigenetic transcriptome microarray identified 416 significantly upregulated and 500 significantly downregulated mRNA peaks in neutrophils of ischemic stroke patients. Moreover, 48 mRNAs and 18 lncRNAs were hypermethylated, and 115 mRNAs and 29 lncRNAs were hypomethylated after cerebral ischemia. Gene ontology (GO) analysis identified that these m6A-modified mRNAs were primarily enriched in calcium ion transport, long-term synaptic potentiation, and base-excision repair. The signaling pathways involved were EGFR tyrosine kinase inhibitor resistance, ErbB, and base excision repair signaling pathway. MeRIP-qPCR validation results showed that NRG1 and GDPD1 were significantly hypermethylated, and LIG1, CHRND, lncRNA RP11-442J17.2, and lncRNA RP11-600P1.2 were significantly hypomethylated after cerebral ischemia. Moreover, the expression levels of major m6A regulators Mettl3, Fto, Ythdf1, and Ythdf3 were obviously declined in the brain and leukocytes of post-stroke mouse models. CONCLUSION This study explored the RNA m6A methylation pattern in the neutrophils of ischemic stroke patients, indicating that it is an intervention target of epigenetic regulation in ischemic stroke.
Collapse
Affiliation(s)
- Junfen Fan
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China; Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing 100053, China.
| | - Liyuan Zhong
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Feng Yan
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China; Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing 100053, China
| | - Xue Li
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Lingzhi Li
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Haiping Zhao
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China; Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing 100053, China
| | - Ziping Han
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China; Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing 100053, China
| | - Rongliang Wang
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China; Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing 100053, China
| | - Zhen Tao
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China; Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing 100053, China
| | - Yangmin Zheng
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China; Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing 100053, China
| | - Qingfeng Ma
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China.
| | - Yumin Luo
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China; Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing 100053, China; Beijing Institute for Brain Disorders, Capital Medical University, Beijing 100053, China.
| |
Collapse
|
4
|
Zhong FF, Wei B, Bao GX, Lou YP, Wei ME, Wang XY, Xiao X, Tian JJ. FABP3 Induces Mitochondrial Autophagy to Promote Neuronal Cell Apoptosis in Brain Ischemia-Reperfusion Injury. Neurotox Res 2024; 42:35. [PMID: 39008165 DOI: 10.1007/s12640-024-00712-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 05/30/2024] [Accepted: 07/05/2024] [Indexed: 07/16/2024]
Abstract
This study elucidates the molecular mechanisms by which FABP3 regulates neuronal apoptosis via mitochondrial autophagy in the context of cerebral ischemia-reperfusion (I/R). Employing a transient mouse model of middle cerebral artery occlusion (MCAO) established using the filament method, brain tissue samples were procured from I/R mice. High-throughput transcriptome sequencing on the Illumina CN500 platform was performed to identify differentially expressed mRNAs. Critical genes were selected by intersecting I/R-related genes from the GeneCards database with the differentially expressed mRNAs. The in vivo mechanism was explored by infecting I/R mice with lentivirus. Brain tissue injury, infarct volume ratio in the ischemic penumbra, neurologic deficits, behavioral abilities, neuronal apoptosis, apoptotic factors, inflammatory factors, and lipid peroxidation markers were assessed using H&E staining, TTC staining, Longa scoring, rotation experiments, immunofluorescence staining, and Western blot. For in vitro validation, an OGD/R model was established using primary neuron cells. Cell viability, apoptosis rate, mitochondrial oxidative stress, morphology, autophagosome formation, membrane potential, LC3 protein levels, and colocalization of autophagosomes and mitochondria were evaluated using MTT assay, LDH release assay, flow cytometry, ROS/MDA/GSH-Px measurement, transmission electron microscopy, MitoTracker staining, JC-1 method, Western blot, and immunofluorescence staining. FABP3 was identified as a critical gene in I/R through integrated transcriptome sequencing and bioinformatics analysis. In vivo experiments revealed that FABP3 silencing mitigated brain tissue damage, reduced infarct volume ratio, improved neurologic deficits, restored behavioral abilities, and attenuated neuronal apoptosis, inflammation, and mitochondrial oxidative stress in I/R mice. In vitro experiments demonstrated that FABP3 silencing restored OGD/R cell viability, reduced neuronal apoptosis, and decreased mitochondrial oxidative stress. Moreover, FABP3 induced mitochondrial autophagy through ROS, which was inhibited by the free radical scavenger NAC. Blocking mitochondrial autophagy with sh-ATG5 lentivirus confirmed that FABP3 induces mitochondrial dysfunction and neuronal apoptosis by activating mitochondrial autophagy. In conclusion, FABP3 activates mitochondrial autophagy through ROS, leading to mitochondrial dysfunction and neuronal apoptosis, thereby promoting cerebral ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Fang-Fang Zhong
- Department of Neurology, Shaoxing People's Hospital, Shaoxing, China.
| | - Bo Wei
- Department of Neurology, Shaoxing People's Hospital, Shaoxing, China
| | - Guo-Xiang Bao
- Department of Clinical Laboratory Center, Shaoxing People's Hospital, Shaoxing, China
| | - Yi-Ping Lou
- Department of Neurology, Shaoxing People's Hospital, Shaoxing, China
| | - Ming-Er Wei
- Department of Neurology, Shaoxing People's Hospital, Shaoxing, China
| | - Xin-Yue Wang
- Department of Neurology, Shaoxing People's Hospital, Shaoxing, China
| | - Xiao Xiao
- School of Medicine, Shaoxing University, Shaoxing, China
| | - Jin-Jin Tian
- School of Medicine, Shaoxing University, Shaoxing, China
| |
Collapse
|
5
|
Chokkalla AK, Arruri V, Mehta SL, Vemuganti R. Loss of Epitranscriptomic Modification N 6-Methyladenosine (m 6A) Reader YTHDF1 Exacerbates Ischemic Brain Injury in a Sexually Dimorphic Manner. Transl Stroke Res 2024:10.1007/s12975-024-01267-4. [PMID: 38869772 DOI: 10.1007/s12975-024-01267-4] [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: 03/28/2024] [Revised: 05/28/2024] [Accepted: 06/06/2024] [Indexed: 06/14/2024]
Abstract
N6-Methyladenosine (m6A) is a neuronal-enriched, reversible post-transcriptional modification that regulates RNA metabolism. The m6A-modified RNAs recruit various m6A-binding proteins that act as readers. Differential m6A methylation patterns are implicated in ischemic brain damage, yet the precise role of m6A readers in propagating post-stroke m6A signaling remains unclear. We presently evaluated the functional significance of the brain-enriched m6A reader YTHDF1, in post-stroke pathophysiology. Focal cerebral ischemia significantly increased YTHDF1 mRNA and protein expression in adult mice of both sexes. YTHDF1-/- male, but not female, mice subjected to transient middle cerebral artery occlusion (MCAO) showed worsened motor function recovery and increased infarction compared to sex-matched YTHDF1+/+ mice. YTHDF1-/- male, but not female, mice subjected to transient MCAO also showed significantly perturbed expression of genes related to inflammation, and increased infiltration of peripheral immune cells into the peri-infarct cortex, compared with sex-matched YTHDF1+/+ mice. Thus, this study demonstrates a sexual dimorphism of YTHDF1 in regulating post-ischemic inflammation and pathophysiology. Hence, post-stroke epitranscriptomic regulation might be sex-dependent.
Collapse
Affiliation(s)
- Anil K Chokkalla
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, 53792, USA
- Cellular and Molecular Pathology Graduate Program, University of Wisconsin, Madison, WI, USA
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Vijay Arruri
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, 53792, USA
| | - Suresh L Mehta
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, 53792, USA
| | - Raghu Vemuganti
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, 53792, USA.
- Cellular and Molecular Pathology Graduate Program, University of Wisconsin, Madison, WI, USA.
- William S. Middleton Memorial Veteran Administration Hospital, Madison, WI, USA.
| |
Collapse
|
6
|
Liu C, Gao Q, Dong J, Cai H. Usf2 Deficiency Promotes Autophagy to Alleviate Cerebral Ischemia-Reperfusion Injury Through Suppressing YTHDF1-m6A-Mediated Cdc25A Translation. Mol Neurobiol 2024; 61:2556-2568. [PMID: 37914905 DOI: 10.1007/s12035-023-03735-8] [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: 07/24/2023] [Accepted: 10/20/2023] [Indexed: 11/03/2023]
Abstract
Autophagy has been involved in protection of ischemia/reperfusion (I/R)-induced injury in many tissues including the brain. The upstream stimulatory factor 2 (Usf2) was proposed as a regulator in aging and degenerative brain diseases; however, the its role in autophagy during cerebral I/R injury remains unclear. Here, the middle cerebral artery occlusion (MCAO) operation was applied to establish an I/R mouse model. We showed that Usf2 was significantly upregulated in I/R-injured brain, accompanied by decreased levels of autophagy. Then, oxygen-glucose deprivation/recovery (OGD/R) treatment was used to establish a cellular I/R model in HT22 neurons, and lentiviral interference vector against Usf2 (LV-sh-Usf2) was used to infect the neurons. Our results showed that Usf2 was significantly upregulated in OGD/R-treated HT22 neurons that displayed an increased level in cell apoptosis and decreased levels in cell viability and autophagy, and interference of Usf2 largely rescued the effects of OGD/R on cell viability, apoptosis, and autophagy, suggesting an important role of Usf2 in neuron autophagy. In the mechanism exploration, we found that, as a transcription factor, Usf2 bound to the promoter of YTHDF1, a famous reader of N6-Methyladenosine (m6A), also induced by OGD/R, and promoted its transcription. Overexpression of YTHDF1 was able to reverse the improvement of Usf2 interference on viability and autophagy of HT22 neurons. Moreover, YTHDF1 suppressed autophagy to induce HT22 cell apoptosis through increasing m6A-mediated stability of Cdc25A, a newly identified autophagy inhibitor. Finally, we demonstrated that interference of Usf2 markedly improved autophagy and alleviated I/R-induced injury in MCAO mice.
Collapse
Affiliation(s)
- Chao Liu
- Department of Vascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China
| | - Qing Gao
- Department of Anesthesia Operation, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China
| | - Jian Dong
- Department of Vascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China
| | - Hui Cai
- Department of Vascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China.
| |
Collapse
|
7
|
Han F. N6-methyladenosine modification in ischemic stroke: Functions, regulation, and therapeutic potential. Heliyon 2024; 10:e25192. [PMID: 38317953 PMCID: PMC10840115 DOI: 10.1016/j.heliyon.2024.e25192] [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: 06/29/2023] [Revised: 12/09/2023] [Accepted: 01/22/2024] [Indexed: 02/07/2024] Open
Abstract
N6-methyladenosine (m6A) modification is the most frequently occurring internal modification in eukaryotic RNAs. By modulating various aspects of the RNA life cycle, it has been implicated in a wide range of pathological and physiological processes associated with human diseases. Ischemic stroke is a major cause of death and disability worldwide with few treatment options and a narrow therapeutic window, and accumulating evidence has indicated the involvement of m6A modifications in the development and progression of this type of stroke. In this review, which provides insights for the prevention and clinical treatment of stroke, we present an overview of the roles played by m6A modification in ischemic stroke from three main perspectives: (1) the association of m6A modification with established risk factors for stroke, including hypertension, diabetes mellitus, hyperlipidemia, obesity, and heart disease; (2) the roles of m6A modification regulators and their functional regulation in the pathophysiological injury mechanisms of stroke, namely oxidative stress, mitochondrial dysfunction, endothelial dysfunction, neuroinflammation, and cell death processes; and (3) the diagnostic and therapeutic potential of m6A regulators in the treatment of stroke.
Collapse
Affiliation(s)
- Fei Han
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, 100730, China
| |
Collapse
|
8
|
Zhao L, Song D, Li T, Li Y, Dang M, Hao Q, Fan H, Lu Z, Lu J, Heyingwang, Wang X, Jian Y, Zhang G. Identification of significant m6A regulators and immune microenvironment characterization in ischemic stroke. Sci Rep 2024; 14:3456. [PMID: 38342932 PMCID: PMC10859379 DOI: 10.1038/s41598-024-53788-5] [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: 08/01/2023] [Accepted: 02/05/2024] [Indexed: 02/13/2024] Open
Abstract
The role of m6A modification in the regulation of the immune microenvironment (IME) of ischemic stroke (IS) is barely known. Thus, we aim to investigate the impact of m6A modification on the IME of IS and its diagnostic value in IS. We comprehensively assessed the m6A modification patterns, the relationship between these modification patterns and the characteristics of the IME. The m6A modification patterns of individual IS sample were quantified by m6Ascore. The performance of m6A phenotype-related genes as potential biomarkers was evaluated by the area under the receiver operating characteristic curve. Experimental validation was also performed by qRT-PCR. Six dysregulated m6A regulators were identified and a classification model consisting of four key m6A regulators (METLL3, RBMX, RBM15B, YTDHF3) could distinguish IS and healthy control samples well. METTL3 and YTHDF3 are closely related to circulating neutrophil abundance. Two distinct m6A modification patterns were determined which differed in immunocyte abundance. We also identified six m6A phenotype-related genes (APOBEC3A, PTMA, FCGR3A, LOC440926, LOC649946, and FTH1L11), and further explored their biological function. Among them, APOBEC3A, FCGR3A, and FTH1L11 were positively associated with neutrophil abundance. APOBEC3A and FCGR3A were stable diagnostic m6A-associated genes in both the discovery and validation cohorts. This study reveals that m6A modification plays a non-negligible role in the formation of a diversified and complex IME in IS. The m6A phenotype-related genes could be diagnostic biomarkers of IS.
Collapse
Affiliation(s)
- Lili Zhao
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xiwulu, Xi'an, 710004, China
| | - Dingli Song
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Tao Li
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xiwulu, Xi'an, 710004, China
| | - Ye Li
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xiwulu, Xi'an, 710004, China
| | - Meijuan Dang
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xiwulu, Xi'an, 710004, China
| | - Qian Hao
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, Shaanxi, China
| | - Hong Fan
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xiwulu, Xi'an, 710004, China
| | - Ziwei Lu
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xiwulu, Xi'an, 710004, China
| | - Jialiang Lu
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xiwulu, Xi'an, 710004, China
| | - Heyingwang
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xiwulu, Xi'an, 710004, China
| | - Xiaoya Wang
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xiwulu, Xi'an, 710004, China
| | - Yating Jian
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xiwulu, Xi'an, 710004, China
| | - Guilian Zhang
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xiwulu, Xi'an, 710004, China.
| |
Collapse
|
9
|
Liu C, Chen H, Tao X, Li C, Li A, Wu W. ALKBH5 protects against stroke by reducing endoplasmic reticulum stress-dependent inflammation injury via the STAT5/PERK/EIF2α/CHOP signaling pathway in an m 6A-YTHDF1-dependent manner. Exp Neurol 2024; 372:114629. [PMID: 38056583 DOI: 10.1016/j.expneurol.2023.114629] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 10/31/2023] [Accepted: 11/27/2023] [Indexed: 12/08/2023]
Abstract
BACKGROUND Endoplasmic reticulum (ER) stress causes neuroinflammation and neuronal apoptosis during ischemic stroke progression. This study has investigated the role of ALKBH5 in ER stress during ischemic stroke progression. METHODS In vivo and in vitro models of ischemic stroke were established by middle cerebral artery occlusion (MCAO) and OGD/R treatment, respectively. Cerebral infarct size was detected using triphenyltetrazolium chloride staining (TTC), and pathological changes were examined using histological staining. The levels of inflammatory factors were analyzed using Enzyme-linked immunosorbent assay. Cell counting kit-8 assay and flow cytometry were used to measure cell viability and apoptosis, respectively. The global m6A level was detected using the commercial kit, and STAT5 mRNA m6A level was determined using methylated RNA binding protein immunoprecipitation (Me-RIP). ALKBH5, YTHDF1, and STAT5 interactions were analyzed using RIP and RNA pull-down assays. RESULTS ALKBH5 was upregulated in MCAO animals and OGD/R cell models. ALKBH5 knockdown exacerbated ER stress, neuroinflammation, and neuronal apoptosis in brain tissues and neuronal cells. ALKBH5 inhibited STAT5 mRNA stability and expression in an m6A-YTHDF1-dependent manner. STAT5 promoted ER stress by activating the PERK/eIF2/CHOP signaling pathway. Furthermore, STAT5 knockdown reversed the effects of ALKBH5 knockdown on OGD/R-induced ER stress and neuroinflammation in HT22 cells. CONCLUSION ALKBH5 knockdown exacerbated ischemic stroke by increasing ER stress-dependent neuroinflammation and neuronal apoptosis via the STAT5/PERK/EIF2α/CHOP signaling pathway in an m6A-YTHDF1-dependent manner.
Collapse
Affiliation(s)
- Chujuan Liu
- Department of Rehabilitation, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, Guangdong Province, PR China; Department of Rehabilitation, Hunan Provincial People's Hospital, The First Affifiliated Hospital of Hunan Normal University, Changsha 410006, Hunan Province, PR China
| | - Hui Chen
- Department of Rehabilitation, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, Guangdong Province, PR China
| | - Xi Tao
- Department of Rehabilitation, Hunan Provincial People's Hospital, The First Affifiliated Hospital of Hunan Normal University, Changsha 410006, Hunan Province, PR China
| | - Chen Li
- Department of Rehabilitation, Hunan Provincial People's Hospital, The First Affifiliated Hospital of Hunan Normal University, Changsha 410006, Hunan Province, PR China
| | - Aiping Li
- Department of Neurological Neurology, Hunan Provincial People's Hospital, The First Affifiliated Hospital of Hunan Normal University, Changsha 410006, Hunan Province, PR China
| | - Wen Wu
- Department of Rehabilitation, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, Guangdong Province, PR China.
| |
Collapse
|
10
|
Wan X, Ge Y, Xu S, Feng Y, Zhu Y, Yin L, Pu Y, Liang G. m 6A modification and its role in neural development and neurological diseases. Epigenomics 2023; 15:819-833. [PMID: 37718929 DOI: 10.2217/epi-2023-0190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2023] Open
Abstract
N6-methyladenosine (m6A) methylation, the most prevalent post-transcriptional modification in eukaryotes, represents a highly dynamic and reversible process that is regulated by m6A methyltransferases, m6A demethylases and RNA-binding proteins during RNA metabolism, which affects RNA function. Notably, m6A modification is significantly enriched in the brain and exerts regulatory roles in neurogenesis and neurodevelopment through various mechanisms, further influencing the occurrence and progression of neurological disorders. This study systematically summarizes and discusses the latest findings on common m6A regulators, examining their expression, function and mechanisms in neurodevelopment and neurological diseases. Additionally, we explore the potential of m6A modification in diagnosing and treating neurological disorders, aiming to provide new insights into the molecular mechanisms and potential therapeutic strategies for neurological disorders.
Collapse
Affiliation(s)
- Xin Wan
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, 210009, PR China
| | - Yiling Ge
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, 210009, PR China
| | - Siyi Xu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, 210009, PR China
| | - Yanlu Feng
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, 210009, PR China
| | - Yuxin Zhu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, 210009, PR China
| | - Lihong Yin
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, 210009, PR China
| | - Yuepu Pu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, 210009, PR China
| | - Geyu Liang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, 210009, PR China
| |
Collapse
|
11
|
Lv J, Xing L, Zhong X, Li K, Liu M, Du K. Role of N6-methyladenosine modification in central nervous system diseases and related therapeutic agents. Biomed Pharmacother 2023; 162:114583. [PMID: 36989722 DOI: 10.1016/j.biopha.2023.114583] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/14/2023] [Accepted: 03/21/2023] [Indexed: 03/29/2023] Open
Abstract
N6-methyladenosine (m6A) is a ubiquitous mRNA modification in eukaryotes. m6A occurs through the action of methyltransferases, demethylases, and methylation-binding proteins. m6A methylation of RNA is associated with various neurological disorders, including Alzheimer's disease (AD), Parkinson's disease (PD), depression, cerebral apoplexy, brain injury, epilepsy, cerebral arteriovenous malformations, and glioma. Furthermore, recent studies report that m6A-related drugs have attracted considerable concerns in the therapeutic areas of neurological disorders. Here, we mainly summarized the role of m6A modification in neurological diseases and the therapeutic potential of m6A-related drugs. The aim of this review is expected to be useful to systematically assess m6A as a new potential biomarker and develop innovative modulators of m6A for the amelioration and treatment of neurological disorders.
Collapse
Affiliation(s)
- Junya Lv
- School of Pharmacy, Department of Pharmacology, China Medical University, Shenyang 110122, China
| | - Lijuan Xing
- Precision Laboratory of Panjin Central Hospital, Panjin 124000, China
| | - Xin Zhong
- School of Pharmacy, Department of Pharmacology, China Medical University, Shenyang 110122, China
| | - Kai Li
- Department of Surgical Oncology and General Surgery, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, the First Affiliated Hospital of China Medical University, Shenyang 110001, China.
| | - Mingyan Liu
- School of Pharmacy, Department of Pharmacology, China Medical University, Shenyang 110122, China; Liaoning Medical Diagnosis and Treatment Center, Shenyang 110179, China.
| | - Ke Du
- School of Pharmacy, Department of Pharmacology, China Medical University, Shenyang 110122, China; Department of Surgical Oncology and General Surgery, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, the First Affiliated Hospital of China Medical University, Shenyang 110001, China; Liaoning Medical Diagnosis and Treatment Center, Shenyang 110179, China.
| |
Collapse
|