1
|
Su W, Deng J, Yang L, Wang Y, Gong X, Li J. Melatonin alleviates LPS-induced depression-like behavior in mice by inhibiting ferroptosis by regulating RNA methylation-mediated SIRT6/Nrf2/HO-1 pathway. Eur J Nutr 2024; 63:3133-3148. [PMID: 39294335 DOI: 10.1007/s00394-024-03495-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 08/23/2024] [Indexed: 09/20/2024]
Abstract
PURPOSE The objective of this study is to investigate the impact of ferroptosis on depression and elucidate the molecular mechanism underlying melatonin's inhibitory effect on ferroptosis in the treatment of depression. METHODS In this study, a depression-like behavior model was induced in mice using LPS, and the effect of melatonin on depression-like behavior was evaluated through behavioral experiments (such as forced swimming test (FST) and sucrose preference test (SPT)). Additionally, molecular biological techniques (including real-time fluorescence quantitative PCR, Western blotting, immunoprecipitation) were employed to detect the expression levels and interactions of METTL3, SIRT6 and ferroptosis-related genes in mouse brain tissue. Furthermore, both in vitro and in vivo experiments were conducted to verify the regulatory effect of melatonin on Nrf2/HO-1 pathway and explore its potential molecular mechanism for regulating ferroptosis. RESULTS Melatonin was found to significantly ameliorate depression-like behavior in mice, as evidenced by reduced immobility time in the forced swimming test and increased sucrose intake in the sucrose preference test. Subsequent investigations revealed that melatonin modulated SIRT6 stability through METTL3-mediated ubiquitination of SIRT6, leading to its degradation. As a deacetylase, SIRT6 plays a pivotal role in cellular metabolism regulation and antioxidative stress response. This study elucidated potential signaling pathways involving Nrf2/HO-1 through which SIRT6 may exert its effects. CONCLUSION The findings suggest that melatonin can improve depressive behavior by suppressing ferroptosis and protecting neurons through its antioxidant properties. Additionally, targeting the Nrf2/HO-1 pathway via METTL3 and NEDD4 regulation may be a potential therapeutic approach for depression.
Collapse
Affiliation(s)
- Wenjie Su
- 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, Sichuan, 610072, China
| | - Jia Deng
- 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, Sichuan, 610072, China
| | - Lina Yang
- 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, Sichuan, 610072, China
| | - Yu Wang
- 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, Sichuan, 610072, China
| | - Xinran Gong
- 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, Sichuan, 610072, China.
| | - Jiacen Li
- 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, Sichuan, 610072, China.
| |
Collapse
|
2
|
Zhao S, Dong Y, Li Y, Wang Z, Chen Y, Dong Y. Melatonin Alleviates Lipopolysaccharide-Induced Abnormal Pregnancy through MTNR1B Regulation of m6A. Int J Mol Sci 2024; 25:733. [PMID: 38255808 PMCID: PMC10815701 DOI: 10.3390/ijms25020733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 12/18/2023] [Accepted: 01/02/2024] [Indexed: 01/24/2024] Open
Abstract
Pregnancy is a highly intricate and delicate process, where inflammation during early stages may lead to pregnancy loss or defective implantation. Melatonin, primarily produced by the pineal gland, exerts several pharmacological effects. N6-methyladenosine (m6A) is the most prevalent mRNA modification in eukaryotes. This study aimed to investigate the association between melatonin and m6A during pregnancy and elucidate the underlying protective mechanism of melatonin. Melatonin was found to alleviate lipopolysaccharide (LPS)-induced reductions in the number of implantation sites. Additionally, it mitigated the activation of inflammation, autophagy, and apoptosis pathways, thereby protecting the pregnancy process in mice. The study also revealed that melatonin regulates uterine m6A methylation levels and counteracts abnormal changes in m6A modification of various genes following LPS stimulation. Furthermore, melatonin was shown to regulate m6A methylation through melatonin receptor 1B (MTNR1B) and subsequently modulate inflammation, autophagy, and apoptosis through m6A. In conclusion, our study demonstrates that melatonin protects pregnancy by influencing inflammation, autophagy, and apoptosis pathways in an m6A-dependent manner via MTNR1B. These findings provide valuable insights into the mechanisms underlying melatonin's protective effects during pregnancy and may have implications for potential therapeutic strategies in managing pregnancy-related complications.
Collapse
Affiliation(s)
- Shisu Zhao
- Laboratory of Neurobiology, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (S.Z.); (Y.D.); (Y.L.); (Z.W.)
| | - Yanjun Dong
- Laboratory of Neurobiology, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (S.Z.); (Y.D.); (Y.L.); (Z.W.)
| | - Yuanyuan Li
- Laboratory of Neurobiology, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (S.Z.); (Y.D.); (Y.L.); (Z.W.)
| | - Zixu Wang
- Laboratory of Neurobiology, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (S.Z.); (Y.D.); (Y.L.); (Z.W.)
| | - Yaoxing Chen
- Laboratory of Neurobiology, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (S.Z.); (Y.D.); (Y.L.); (Z.W.)
| | - Yulan Dong
- Laboratory of Neurobiology, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (S.Z.); (Y.D.); (Y.L.); (Z.W.)
- Key Laboratory of Precision Nutrition and Food Quality, Ministry of Education, China Agricultural University, Beijing 100193, China
| |
Collapse
|
3
|
Gan Z, Zhao M, Xia Y, Yan Y, Ren W. Carbon metabolism in the regulation of macrophage functions. Trends Endocrinol Metab 2024; 35:62-73. [PMID: 37778898 DOI: 10.1016/j.tem.2023.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 09/07/2023] [Accepted: 09/08/2023] [Indexed: 10/03/2023]
Abstract
Carbon metabolism, including one-carbon (1C) metabolism and central carbon metabolism (CCM), provides energy for the cell and generates metabolites with signaling activities. The regulation of macrophage polarization involves complex signals and includes an epigenetic level. Epigenetic modifications through changes in carbon metabolism allow macrophages to respond in a timely manner to their environment and adapt to metabolic demands during macrophage polarization. Here we summarize the current understanding of the crosstalk between carbon metabolism and epigenetic modifications in macrophages under physiological conditions and in the tumor microenvironment (TME) and provide targets and further directions for macrophage-associated diseases.
Collapse
Affiliation(s)
- Zhending Gan
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510000, Guangdong, China
| | - Muyang Zhao
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510000, Guangdong, China
| | - Yaoyao Xia
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| | - Yuqi Yan
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Wenkai Ren
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510000, Guangdong, China.
| |
Collapse
|
4
|
Liao X, Chen L, Liu J, Hu H, Hou D, You R, Wang X, Huang H. m 6A RNA methylation regulators predict prognosis and indicate characteristics of tumour microenvironment infiltration in acute myeloid leukaemia. Epigenetics 2023; 18:2160134. [PMID: 36567510 PMCID: PMC9980463 DOI: 10.1080/15592294.2022.2160134] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Patients with acute myeloid leukaemia (AML) have poor prognoses and low overall survival (OS) rates owing to its heterogeneity and the complexity of its tumour microenvironment (TME). N6-methyladenosine (m6A) modification plays a key role in the initiation and progression of haematopoietic malignancies. However, the underlying function of m6A regulators in AML remains elusive. This study thoroughly analysed the m6A modification features of 177 AML patients based on 22 m6A regulators. Utilizing unsupervised clustering, we determined three distinct m6A modification patterns related to different biological functions, TME cell-infiltrating characteristics and clinical outcomes. Additionally, a risk score was constructed based on six m6A regulators-associated prognostic signatures and was validated as an independent and valuable prognostic factor for AML. Patients with a low-risk score exhibited better survival than those with a high-risk score. Many m6A regulators were aberrantly expressed in AML, among which METTL14, YTHDC2, ZC3H13 and RBM15 were observed to be associated with the OS of AML. In addition, these four m6A regulators were found to be noticeably related to the immune checkpoint inhibitor (ICI) treatments. Finally, we verified the expression levels of these four m6A regulators in AML and healthy samples and three groups of AML patients with different risk categories. Collectively, our study indicates that the m6A modification pattern is involved in TME immune-infiltrating characteristics and prognosis in AML. A better understanding of the m6A modification pattern will help enhance our knowledge of the molecular mechanisms of AML and develop potential prognosis prediction indicators and more effective immunotherapeutic strategies.
Collapse
Affiliation(s)
- Xinai Liao
- Central Laboratory, Fujian Medical University Union Hospital, Fuzhou, China
| | - Ling Chen
- Central Laboratory, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jingru Liu
- Central Laboratory, Fujian Medical University Union Hospital, Fuzhou, China
| | - Haoran Hu
- Central Laboratory, Fujian Medical University Union Hospital, Fuzhou, China
| | - Diyu Hou
- Central Laboratory, Fujian Medical University Union Hospital, Fuzhou, China
| | - Ruolan You
- Central Laboratory, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xiaoting Wang
- Central Laboratory, Fujian Medical University Union Hospital, Fuzhou, China
| | - Huifang Huang
- Central Laboratory, Fujian Medical University Union Hospital, Fuzhou, China
| |
Collapse
|
5
|
Meng Y, Yang W, Li J, Chai W. KIAA1429 facilitates progression of hepatocellular carcinoma by modulating m6A levels in HPN. Heliyon 2023; 9:e22084. [PMID: 38058614 PMCID: PMC10695992 DOI: 10.1016/j.heliyon.2023.e22084] [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: 06/06/2023] [Revised: 11/01/2023] [Accepted: 11/03/2023] [Indexed: 12/08/2023] Open
Abstract
Background Most N6-methyladenosine (m6A)-associated modulatory proteins are involved in the pathogenesis of various cancers. The roles of m6A-related genes in liver hepatocellular carcinoma (LIHC) and the associated mechanisms remain unknown. Methods GEO and GEPIA2 databases were used to identify the m6A modification-related genes which were differentially expressed in LIHC and adjacent non-tumor tissues, and quantitative PCR was used to evaluate the expression of KIAA1429, a major m6A methyltransferase, in LIHC cells. The effect of KIAA1429 on the malignant phenotypes of LIHC cells was evaluated in vitro. The UALCAN, GEPIA, and GEO databases and western blotting assays were used to identify the target genes of KIAA1429. Results KIAA1429 expression was markedly elevated in LIHC tissues, and patients with LIHC who had high KIAA1429 expression had a worse prognosis than those who had low expression. KIAA1429 silencing attenuated LIHC metastasis and proliferation. KIAA142 regulates m6A levels in HPN to intensify LIHC progression. Conclusion Our study suggests a KIAA1429-HPN modulatory model based on m6A modifications, that offers insights into the occurrence and development of LIHC.
Collapse
Affiliation(s)
- Yu Meng
- The First Department of Hepatobiliary and Pancreatic Surgery, Cangzhou Central Hospital, Cangzhou, China
| | - Wenwen Yang
- The Department of Nursing, Cangzhou Medical College, Cangzhou, China
| | - Jinchao Li
- The First Department of Hepatobiliary and Pancreatic Surgery, Cangzhou Central Hospital, Cangzhou, China
| | - Wei Chai
- The First Department of Hepatobiliary and Pancreatic Surgery, Cangzhou Central Hospital, Cangzhou, China
| |
Collapse
|
6
|
Zou K, Dong H, Li M, Zhang Y, Zhang K, Song D, Chu C. Comprehensive analysis of transcriptome-wide N6-methyladenosine methylomes in the Barrett's esophagus in rats. Genomics 2023; 115:110687. [PMID: 37454940 DOI: 10.1016/j.ygeno.2023.110687] [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: 05/02/2023] [Revised: 06/22/2023] [Accepted: 07/13/2023] [Indexed: 07/18/2023]
Abstract
PURPOSE As the most abundant RNA modification, N6-methyladenosine (m6A) methylation plays crucial roles in various diseases. The aim of this study is to comprehensively map the landscape of the mRNA m6A modification pattern in Barrett's esophagus (BE) in order to find key genes and potential therapy for BE and even esophageal adenocarcinoma (EAC). METHODS Methylated RNA immunoprecipitation sequencing (MeRIP-seq) and RNA-sequencing (RNA-seq) were performed to compare the difference in mRNA m6A methylation and differentially expressed mRNAs between BE and normal control (NC) tissues. Bioinformatics analysis was used to describe the m6A modification pattern and specific genes in BE and NC tissues. RESULTS Through MeRIP-seq, we obtained m6A methylation profiling in BE and NC tissues. In total, 11,026 unique peaks were detected in the BE groups, whereas 8564 unique peaks were detected in the NC groups. Peaks were primarily enriched within CDS with GGACU motifs and most of the peaks were within 1000 bp in width. Moreover, functional enrichment analysis demonstrated that hypermethylated and hypomethylated genes were significantly enriched in coronavirus disease pathway, calcium signaling pathway and MAPK signaling pathways. Furthermore, PPI network was conducted and 18 hub genes were identified via STRING database and Cystoscope. Among them, ACTA1, CDC20, CKM, KIF20a, MYH11, TPM2, MYL9, DES, TNNT3 were overexpressed in EAC in the GEPIA gene bank and TPM1, KIF20a impaired patients' survival in the Kaplan-Meier plotter database. Finally, functional enrichment analysis demonstrated that co-expressed genes of TPM1 were significantly enriched in calcium signaling pathway, cGMP-PKG signaling pathway and PI3K-Akt signaling pathway. CONCLUSION Our study is the first to perform comprehensive and transcriptome-wide maps to identify the potential roles played by m6A methylation in BE, which widely involved in oxidative stress. This foresees a guiding role in revealing the molecular mechanism of m6A-mediated genes that govern the pathogenesis and progression of BE and EAC.
Collapse
Affiliation(s)
- Ke Zou
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250013, China; Jinan Central Hospital, Shandong University, No. 105, Jiefang Road, Jinan, Shandong 250013, China; Jinan Digestive Diseases Clinical Research Center, No. 105, Jiefang Road, Jinan, Shandong 250013, China; Jinan Key Translational Research Laboratory in Gastroenterology, No. 105, Jiefang Road, Jinan, Shandong 250013, China; Shandong University, No. 44, Wenhua west Road, Jinan, Shandong 250102, China
| | - Hui Dong
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250013, China; Jinan Central Hospital, Shandong University, No. 105, Jiefang Road, Jinan, Shandong 250013, China; Shandong University, No. 44, Wenhua west Road, Jinan, Shandong 250102, China
| | - Mengmeng Li
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250013, China; Jinan Central Hospital, Shandong University, No. 105, Jiefang Road, Jinan, Shandong 250013, China; Shandong University, No. 44, Wenhua west Road, Jinan, Shandong 250102, China
| | - Ying Zhang
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250013, China
| | - Kai Zhang
- Department of Internal Medicine, Weifang Medical University, Weifang, China
| | - Danlin Song
- Department of Internal Medicine, Weifang Medical University, Weifang, China
| | - Chuanlian Chu
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250013, China; Jinan Central Hospital, Shandong University, No. 105, Jiefang Road, Jinan, Shandong 250013, China; Jinan Digestive Diseases Clinical Research Center, No. 105, Jiefang Road, Jinan, Shandong 250013, China; Jinan Key Translational Research Laboratory in Gastroenterology, No. 105, Jiefang Road, Jinan, Shandong 250013, China; Shandong University, No. 44, Wenhua west Road, Jinan, Shandong 250102, China.
| |
Collapse
|
7
|
Zhang T, Zhang L, Huang G, Hao X, Liu Z, Huo S. MEL regulates miR-21 and let-7b through the STAT3 cascade in the follicular granulosa cells of Tibetan sheep. Theriogenology 2023; 205:114-129. [PMID: 37120893 DOI: 10.1016/j.theriogenology.2023.04.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 03/21/2023] [Accepted: 04/10/2023] [Indexed: 05/02/2023]
Abstract
Under physiological and pathological conditions, melatonin (MEL) can regulate microRNA (miRNA) expression. However, the mechanisms underlying the regulatory effects of MEL on miRNAs in ovaries are not understood. Firstly, by using fluorescence in situ hybridisation, we found that in ovaries and follicular granulosa cells (FGCs), MT1 co-located with miR-21 and let-7b. Additionally, immunofluorescence revealed that MT1, STAT3, c-MYC and LIN28 proteins co-located. The mRNA and protein levels of STAT3, c-MYC and LIN28 increased under treatment with 10-7 M MEL. MEL induced an increase in miR-21 and a decrease in let-7b. The LIN28/let-7b and STAT3/miR-21 axes are related to cell differentiation, apoptosis and proliferation. We explored whether the STAT3/c-MYC/LIN28 pathway was involved in miRNA regulation by MEL to explore the putative mechanism of the above relationship. AG490, an inhibitor of the STAT3 pathway, was added before MEL treatment. AG490 inhibited the MEL-induced increases in STAT3, c-MYC, LIN28 and MT1 and changes in miRNA. Through live-cell detection, we discovered that MEL enhanced the proliferation of FGCs. However, the ki67 protein levels decreased when AG490 was added in advance. Furthermore, the dual-luciferase reporter assay verified that STAT3, LIN28 and MT1 were target genes of let-7b. Furthermore, STAT3 and SMAD7 were target genes of miR-21. In addition, the protein levels of the STAT3, c-MYC, LIN28 and MEL receptors decreased when let-7b was overexpressed in FGCs. Overall, MEL might regulate miRNA expression through the STAT3 pathway. In addition, a negative feedback loop between the STAT3 and miR-21 formed; MEL and let-7b antagonized each other in FGCs. These findings may provide a theoretical basis for improving the reproductive performance of Tibetan sheep through MEL and miRNAs.
Collapse
Affiliation(s)
- Taojie Zhang
- Northwest Minzu University, Life Science and Engineering College, Lanzhou, Gansu, China.
| | - Lijuan Zhang
- Northwest Minzu University, Life Science and Engineering College, Lanzhou, Gansu, China
| | - Guoliang Huang
- Northwest Minzu University, Life Science and Engineering College, Lanzhou, Gansu, China
| | - Xiaomeng Hao
- Northwest Minzu University, Life Science and Engineering College, Lanzhou, Gansu, China
| | - Zezheng Liu
- Northwest Minzu University, Life Science and Engineering College, Lanzhou, Gansu, China
| | - Shengdong Huo
- Northwest Minzu University, Life Science and Engineering College, Lanzhou, Gansu, China.
| |
Collapse
|
8
|
Shukla M, Vincent B. Melatonin as a Harmonizing Factor of Circadian Rhythms, Neuronal Cell Cycle and Neurogenesis: Additional Arguments for Its Therapeutic Use in Alzheimer's Disease. Curr Neuropharmacol 2023; 21:1273-1298. [PMID: 36918783 PMCID: PMC10286584 DOI: 10.2174/1570159x21666230314142505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 12/07/2022] [Accepted: 12/31/2022] [Indexed: 03/16/2023] Open
Abstract
The synthesis and release of melatonin in the brain harmonize various physiological functions. The apparent decline in melatonin levels with advanced aging is an aperture to the neurodegenerative processes. It has been indicated that down regulation of melatonin leads to alterations of circadian rhythm components, which further causes a desynchronization of several genes and results in an increased susceptibility to develop neurodegenerative diseases. Additionally, as circadian rhythms and memory are intertwined, such rhythmic disturbances influence memory formation and recall. Besides, cell cycle events exhibit a remarkable oscillatory system, which is downstream of the circadian phenomena. The linkage between the molecular machinery of the cell cycle and complex fundamental regulatory proteins emphasizes the conjectural regulatory role of cell cycle components in neurodegenerative disorders such as Alzheimer's disease. Among the mechanisms intervening long before the signs of the disease appear, the disturbances of the circadian cycle, as well as the alteration of the machinery of the cell cycle and impaired neurogenesis, must hold our interest. Therefore, in the present review, we propose to discuss the underlying mechanisms of action of melatonin in regulating the circadian rhythm, cell cycle components and adult neurogenesis in the context of AD pathogenesis with the view that it might further assist to identify new therapeutic targets.
Collapse
Affiliation(s)
- Mayuri Shukla
- Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom 73170, Thailand
- Present Address: Chulabhorn Graduate Institute, Chulabhorn Royal Academy, 10210, Bangkok, Thailand
| | - Bruno Vincent
- Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom 73170, Thailand
- Institute of Molecular and Cellular Pharmacology, Laboratory of Excellence DistALZ, Université Côte d'Azur, INSERM, CNRS, Sophia-Antipolis, 06560, Valbonne, France
| |
Collapse
|
9
|
The Intricate Interplay between the ZNF217 Oncogene and Epigenetic Processes Shapes Tumor Progression. Cancers (Basel) 2022; 14:cancers14246043. [PMID: 36551531 PMCID: PMC9776013 DOI: 10.3390/cancers14246043] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/02/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022] Open
Abstract
The oncogenic transcription factor ZNF217 orchestrates several molecular signaling networks to reprogram integrated circuits governing hallmark capabilities within cancer cells. High levels of ZNF217 expression provide advantages to a specific subset of cancer cells to reprogram tumor progression, drug resistance and cancer cell plasticity. ZNF217 expression level, thus, provides a powerful biomarker of poor prognosis and a predictive biomarker for anticancer therapies. Cancer epigenetic mechanisms are well known to support the acquisition of hallmark characteristics during oncogenesis. However, the complex interactions between ZNF217 and epigenetic processes have been poorly appreciated. Deregulated DNA methylation status at ZNF217 locus or an intricate cross-talk between ZNF217 and noncoding RNA networks could explain aberrant ZNF217 expression levels in a cancer cell context. On the other hand, the ZNF217 protein controls gene expression signatures and molecular signaling for tumor progression by tuning DNA methylation status at key promoters by interfering with noncoding RNAs or by refining the epitranscriptome. Altogether, this review focuses on the recent advances in the understanding of ZNF217 collaboration with epigenetics processes to orchestrate oncogenesis. We also discuss the exciting burgeoning translational medicine and candidate therapeutic strategies emerging from those recent findings connecting ZNF217 to epigenetic deregulation in cancer.
Collapse
|
10
|
Guo L, Huai Q, Zhou B, Ying J, Guo W. Comprehensive analysis of the prognostic impact and immune implication of KIAA1429 in lung adenocarcinoma. CANCER INNOVATION 2022; 1:328-343. [PMID: 38089085 PMCID: PMC10686173 DOI: 10.1002/cai2.40] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 11/02/2022] [Accepted: 11/11/2022] [Indexed: 10/15/2024]
Abstract
Background Lung adenocarcinoma (LUAD) is the most common lung cancer worldwide. N6-methyladenosine (m6A) methylation is a messenger RNA (mRNA) modification that plays a key role in tumor growth, immune microenvironment, and immunotherapy response. This study investigated the expression level, mutation status, prognostic value, and predictive ability for response to anti-PD-1 immunotherapy of the m6A methyltransferase KIAA1429 in LUAD. Methods This study examined multiple public data cohorts and independent samples from National Cancer Center (NCC) to evaluate the clinical significance and prognostic value of KIAA1429 in LUAD using bioinformatics techniques and immunohistochemical staining. We also evaluated the predictive value of KIAA1429 expression for anti-PD-1 immunotherapy efficacy. GSEA analysis was performed using KIAA1429 RNA-seq data at the tumor tissue level and cellular level to explore the potential molecular mechanism. Results In public databases, KIAA1429 was significantly associated with clinicopathological parameters in LUAD patients and had the potential to predict patient prognosis. The mutation characteristics of KIAA1429-related genes were analyzed and TP53, TTN, CSMD3, and other genes showed high mutation frequencies in LUAD. An independent cohort of 415 samples confirmed that high KIAA1429 expression was significantly associated with poorer prognosis in LUAD patients. Analysis of a small immunotherapy cohort showed that patients with high expression of KIAA1429 had better response after immunotherapy, and the proportion of patients with immunotherapy response was higher in this group. Conclusions Our study confirmed that KIAA1429 was highly expressed in LUAD and was significantly associated with poor prognosis. Moreover, KIAA1429 may serve as a potential marker to predict the efficacy of immunotherapy in LUAD.
Collapse
Affiliation(s)
- Lei Guo
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingThe People's Republic of China
| | - Qilin Huai
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingThe People's Republic of China
| | - Bolun Zhou
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingThe People's Republic of China
| | - Jianming Ying
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingThe People's Republic of China
| | - Wei Guo
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingThe People's Republic of China
- Key Laboratory of Minimally Invasive Therapy Research for Lung CancerChinese Academy of Medical SciencesBeijingThe People's Republic of China
| |
Collapse
|
11
|
Guan S, He Y, Su Y, Zhou L. A Risk Signature Consisting of Eight m 6A Methylation Regulators Predicts the Prognosis of Glioma. Cell Mol Neurobiol 2022; 42:2733-2743. [PMID: 34432221 DOI: 10.1007/s10571-021-01135-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 07/27/2021] [Indexed: 01/05/2023]
Abstract
Glioma progression seriously correlates to the epigenetic context. This study aims to identify glioma subtypes by clustering analysis of patients using the multi-omics data of N6-methyladenosine (m6A) methylation regulators and to construct a risk signature for investigating the role of m6A methylation regulators in the prognosis of glioma. Multi-omics data of glioma and normal control tissues were obtained through The Cancer Genome Atlas (TCGA) database. The clustering analysis of multi-omics data of patients was conducted using the R package iClusterPlus software. The risk model was constructed by univariate and multivariate Cox analysis, and the glioma expression data and related clinical data were obtained by Chinese Glioma Genome Atlas (CGGA) datasets to verify the risk model. By analyzing the glioma data in TCGA, we found that the risk signature could be constructed according to the eight genes with m6A methylation modification function, including ALKBH5, HNRNPA2B1, IGF2BP2, IGF2BP3, RBM15, WTAP, YTHDF1, and YTHDF2. Meanwhile, we found that IGF2BP2 and IGF2BP3 were highly expressed in glioma subtypes with high-risk scores and closely related to the prognosis of glioma patients. m6A methylation regulators, especially IGF2BP2 and IGF2BP3, play important roles in the malignant progression of glioma. The risk signature constructed by eight m6A methylation regulators can predict the prognosis of glioma. IGF2BP2 and IGF2BP3 may be the key regulatory factors of m6A methylation regulators involved in the occurrence and development of glioma, and can serve as molecular markers for the prognosis of glioma.
Collapse
Affiliation(s)
- Sizhong Guan
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, 110001, People's Republic of China
| | - Ye He
- Department of Laboratory Medicine, The First Hospital of China Medical University, Shenyang, 110001, People's Republic of China
| | - Yanna Su
- Department of Laboratory Medicine, The First Hospital of China Medical University, Shenyang, 110001, People's Republic of China
| | - Liping Zhou
- Post Graduation Training Department, The First Hospital of China Medical University, No. 155, Northern Nanjing Road, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China.
| |
Collapse
|
12
|
Sikorski V, Vento A, Kankuri E. Emerging roles of the RNA modifications N6-methyladenosine and adenosine-to-inosine in cardiovascular diseases. MOLECULAR THERAPY. NUCLEIC ACIDS 2022; 29:426-461. [PMID: 35991314 PMCID: PMC9366019 DOI: 10.1016/j.omtn.2022.07.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Cardiovascular diseases lead the mortality and morbidity disease metrics worldwide. A multitude of chemical base modifications in ribonucleic acids (RNAs) have been linked with key events of cardiovascular diseases and metabolic disorders. Named either RNA epigenetics or epitranscriptomics, the post-transcriptional RNA modifications, their regulatory pathways, components, and downstream effects substantially contribute to the ways our genetic code is interpreted. Here we review the accumulated discoveries to date regarding the roles of the two most common epitranscriptomic modifications, N6-methyl-adenosine (m6A) and adenosine-to-inosine (A-to-I) editing, in cardiovascular disease.
Collapse
Affiliation(s)
- Vilbert Sikorski
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
| | - Antti Vento
- Heart and Lung Center, Helsinki University Hospital, 00029 Helsinki, Finland
| | - Esko Kankuri
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
| | - IHD-EPITRAN Consortium
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
- Heart and Lung Center, Helsinki University Hospital, 00029 Helsinki, Finland
| |
Collapse
|
13
|
Liang Y, Zhang X, Ma C, Hu J. m 6A Methylation Regulators Are Predictive Biomarkers for Tumour Metastasis in Prostate Cancer. Cancers (Basel) 2022; 14:cancers14164035. [PMID: 36011028 PMCID: PMC9406868 DOI: 10.3390/cancers14164035] [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: 07/27/2022] [Revised: 08/16/2022] [Accepted: 08/18/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Recurrence and metastatic progression always lead to dismal outcomes in prostate cancer (PCa). There is no reliable biomarker for the prediction of recurrence and metastasis other than the Prostate Cancer Antigen (PCA). N6-methyladenosine (m6A) is the most common post-transcriptional mRNA modification and is regulated by m6A regulators dynamically. Since m6A modification is associated with cancer development and outgrowth, we performed a consensus clustering on PCa with regard to the gene expression of all m6A regulators. We identified three subtypes of Pca with distinct m6A expression patterns and enriched biological pathways. We also established an m6A score for metastasis prediction based on our clustering, which is potentially a predictive biomarker for Pca metastasis. Abstract Prostate cancer (PCa) is one of the most common cancers in men. Usually, most PCas at initial diagnosis are localized and hormone-dependent, and grow slowly. Patients with localized PCas have a nearly 100% 5-year survival rate; however, the 5-year survival rate of metastatic or progressive PCa is still dismal. N6-methyladenosine (m6A) is the most common post-transcriptional mRNA modification and is dynamically regulated by m6A regulators. A few studies have shown that the abnormal expression of m6A regulators is significantly associated with cancer progression and immune cell infiltration, but the roles of these regulators in PCa remain unclear. Here, we examined the expression profiles and methylation levels of 21 m6A regulators across the Cancer Genome Atlas (TCGA), 495 PCas by consensus clustering, and correlated the expression of m6A regulators with PCa progression and immune cell infiltration. Consensus clustering was applied for subtyping Pca samples into clusters based on the expression profiles of m6A regulators. Each subtype’s signature genes were obtained by a pairwise differential expression analysis. Featured pathways of m6A subtypes were predicted by Gene Ontology. The m6A score was developed to predict m6A activation. The association of the m6A score with patients’ survival, metastasis and immune cell infiltration was also investigated. We identified three distinct clusters in PCa based on the expression profiles of 21 m6A regulators by consensus clustering. The differential expression and pathway analyses on the three clusters uncovered the m6A regulators involved in metabolic processes and immune responses in PCa. Moreover, we developed an m6A score to evaluate the m6A regulator activation for PCa. The m6A score is significantly associated with Gleason scores and metastasis in PCa. The predictive capacity of the m6A score on PCa metastasis was also validated in another independent cohort with an area under the curve of 89.5%. Hence, our study revealed the critical role of m6A regulators in PCa progression and the m6A score is a promising predictive biomarker for PCa metastasis.
Collapse
Affiliation(s)
- Yingchun Liang
- Department of Urology, Huashan Hospital, Fudan University, No. 12 WuLuMuQi Middle Road, Shanghai 200040, China
- Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Xiaohua Zhang
- Department of Urology, Huashan Hospital, Fudan University, No. 12 WuLuMuQi Middle Road, Shanghai 200040, China
| | - Chenkai Ma
- Molecular Diagnostic Solution, Nutrition and Health, Health and Biosecurity, CSIRO, North Ryde 2113, Australia
- Correspondence: (C.M.); (J.H.)
| | - Jimeng Hu
- Department of Urology, Huashan Hospital, Fudan University, No. 12 WuLuMuQi Middle Road, Shanghai 200040, China
- Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai 200040, China
- Correspondence: (C.M.); (J.H.)
| |
Collapse
|
14
|
Xu CL, Tan QY, Yang H, Li CY, Wu Z, Ma YF. Melatonin enhances spermatogonia activity through promoting KIAA1429-mediated m 6A deposition to activate the PI3K/AKT signaling. Reprod Biol 2022; 22:100681. [PMID: 35987159 DOI: 10.1016/j.repbio.2022.100681] [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: 04/17/2022] [Revised: 07/28/2022] [Accepted: 07/29/2022] [Indexed: 11/19/2022]
Abstract
Melatonin is a key neuroendocrine hormone that promotes spermatogenesis and sperm motility, but the underlying mechanisms remains poorly understood. In this study, we aimed to investigate the possible roles of m6A (N6--methyl-adenosine) in mediating melatonin-regulated spermatogonia activity alterations. In this study, mouse-derived GC-1 spermatogonia (spg) cell line was used as the in vitro cellular model. The viability, proliferation rates and apoptosis of spermatogonia were detected via CCK-8, Edu staining and flow cytometry respectively. Total m6A level was quantitated by dot blot, while mRNA and proteins contents in spermatogonia were measured by qRT-PCR and western blot respectively. Differentially expressed mRNAs were characterized by deep RNA sequencing method. Results showed that melatonin significantly promoted viability and proliferation rate while inhibited apoptosis in the GC-1 spg cells. The total m6A levels in GC-1 spg cells were also greatly increased by melatonin treatment, accompanied by remarkable expressional elevation of the m6A writer KIAA1429. Moreover, the regulation of GC-1 spg cell viability, proliferation and apoptosis by melatonin were greatly abrogated by KIAA1429 silencing but effectively strengthened by KIAA1429 overexpression. In addition, KIAA1429 overexpression regulates multiple biological process and signaling pathways in spermatogonia such as the PI3K/AKT signaling. The PI3K inhibitor LY294002 effectively mitigated the regulation of spermatogonia activity by KIAA1429 overexpression under melatonin treatment. Taken together, melatonin promotes spermatogonia activity via enhancing KIAA1429 expression and m6A RNA methylation to activate the downstream PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Chang-Long Xu
- Department of Reproductive Medical Center, Nanning Second People Hospital, Nanning 530031, China; National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, Guangxi Medical University, Nanning, Guangxi, 530021, China.
| | - Qing-Ying Tan
- Department of Reproductive Medical Center, Nanning Second People Hospital, Nanning 530031, China.
| | - Hua Yang
- Department of Reproductive Medical Center, Nanning Second People Hospital, Nanning 530031, China
| | - Chun-Yuan Li
- Department of Reproductive Medical Center, Nanning Second People Hospital, Nanning 530031, China
| | - Zhuo Wu
- Department of Reproductive Medical Center, Nanning Second People Hospital, Nanning 530031, China
| | - Ya-Feng Ma
- Department of Obstetrics and Gynecology, Wuxiang Hospital of Nanning Second People Hospital, Nanning 530031,China
| |
Collapse
|
15
|
Loh D, Reiter RJ. Melatonin: Regulation of Viral Phase Separation and Epitranscriptomics in Post-Acute Sequelae of COVID-19. Int J Mol Sci 2022; 23:8122. [PMID: 35897696 PMCID: PMC9368024 DOI: 10.3390/ijms23158122] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 07/09/2022] [Accepted: 07/20/2022] [Indexed: 01/27/2023] Open
Abstract
The relentless, protracted evolution of the SARS-CoV-2 virus imposes tremendous pressure on herd immunity and demands versatile adaptations by the human host genome to counter transcriptomic and epitranscriptomic alterations associated with a wide range of short- and long-term manifestations during acute infection and post-acute recovery, respectively. To promote viral replication during active infection and viral persistence, the SARS-CoV-2 envelope protein regulates host cell microenvironment including pH and ion concentrations to maintain a high oxidative environment that supports template switching, causing extensive mitochondrial damage and activation of pro-inflammatory cytokine signaling cascades. Oxidative stress and mitochondrial distress induce dynamic changes to both the host and viral RNA m6A methylome, and can trigger the derepression of long interspersed nuclear element 1 (LINE1), resulting in global hypomethylation, epigenetic changes, and genomic instability. The timely application of melatonin during early infection enhances host innate antiviral immune responses by preventing the formation of "viral factories" by nucleocapsid liquid-liquid phase separation that effectively blockades viral genome transcription and packaging, the disassembly of stress granules, and the sequestration of DEAD-box RNA helicases, including DDX3X, vital to immune signaling. Melatonin prevents membrane depolarization and protects cristae morphology to suppress glycolysis via antioxidant-dependent and -independent mechanisms. By restraining the derepression of LINE1 via multifaceted strategies, and maintaining the balance in m6A RNA modifications, melatonin could be the quintessential ancient molecule that significantly influences the outcome of the constant struggle between virus and host to gain transcriptomic and epitranscriptomic dominance over the host genome during acute infection and PASC.
Collapse
Affiliation(s)
- Doris Loh
- Independent Researcher, Marble Falls, TX 78654, USA;
| | - Russel J. Reiter
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX 78229, USA
| |
Collapse
|
16
|
Song W, Fei F, Qiao F, Weng Z, Yang Y, Cao B, Yue J, Xu J, Zheng M, Li J. ALKBH5-mediated N 6-methyladenosine modification of TRERNA1 promotes DLBCL proliferation via p21 downregulation. Cell Death Dis 2022; 8:25. [PMID: 35031597 PMCID: PMC8760254 DOI: 10.1038/s41420-022-00819-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 12/09/2021] [Accepted: 12/23/2021] [Indexed: 12/26/2022]
Abstract
Long noncoding RNAs (lncRNAs) have crucial functions in the tumorigenesis and metastasis of cancers. N6-methyladenosine (m6A) modification of RNA is an important epigenetic regulatory mechanism in various malignancies. Nevertheless, the mechanism of m6A-modified lncRNA in diffuse large B cell lymphoma (DLBCL) has remained poorly defined. In the present study, we showed that lncRNA TRERNA1 was associated with the poor prognosis of DLBCL patients. TRERNA1 with internal m6A modification was highly correlated with the demethylase ALKBH5 expression. We further demonstrated that TRERNA1 was a potential downstream target of ALKBH5-mediated m6A modification by m6A-RNA sequencing and m6A-RIP assays. Decreased m6A methylation of TRERNA1 regulated by ALKBH5 was shown to regulate cell proliferation in vitro and in vivo. The results of mechanism analyses revealed that TRERNA1 recruited EZH2 to epigenetically silence the expression of the cyclin-dependent kinases inhibitor p21 by H3K27me3 modification of its promoter region. In addition, ALKBH5 further inhibited p21 expression. Taken together, our results elucidate the functional roles and epigenetic alterations of TRERNA1 through m6A modification in DLBCL. TRERNA1, the expression of which is upregulated by ALKBH5, acts as a scaffold that decreases p21 expression. The results of the present study provide novel targets for the diagnosis and treatment of DLBCL.
Collapse
Affiliation(s)
- Wei Song
- Phase I Clinical Trials Unit, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China. .,Anhui Province Key Laboratory of Translational Cancer Research (Bengbu Medical College), Bengbu, 233030, China.
| | - Fei Fei
- Phase I Clinical Trials Unit, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Fengchang Qiao
- Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health care Hospital, Nanjing, 210001, China
| | - Zuyi Weng
- Phase I Clinical Trials Unit, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Yuanxun Yang
- Phase I Clinical Trials Unit, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Bei Cao
- Phase I Clinical Trials Unit, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Jing Yue
- Department of Gynaecology and Obstetrics, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Jiaxuan Xu
- Phase I Clinical Trials Unit, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Meihong Zheng
- Phase I Clinical Trials Unit, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Juan Li
- Phase I Clinical Trials Unit, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China.
| |
Collapse
|
17
|
Liu W, Tang P, Wang J, Ye W, Ge X, Rong Y, Ji C, Wang Z, Bai J, Fan J, Yin G, Cai W. Extracellular vesicles derived from melatonin-preconditioned mesenchymal stem cells containing USP29 repair traumatic spinal cord injury by stabilizing NRF2. J Pineal Res 2021; 71:e12769. [PMID: 34562326 DOI: 10.1111/jpi.12769] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 09/23/2021] [Accepted: 09/23/2021] [Indexed: 12/27/2022]
Abstract
Spinal cord injury (SCI) is a devastating trauma that leads to irreversible motor and sensory dysfunction and is, so far, without effective treatment. Recently, however, nano-sized extracellular vesicles derived from preconditioned mesenchymal stem cells (MSCs) have shown great promise in treating various diseases, including SCI. In this study, we investigated whether extracellular vesicles (MEVs) derived from MSCs pretreated with melatonin (MT), which is well recognized to be useful in treating diseases, including Alzheimer's disease, non-small cell lung cancer, acute ischemia-reperfusion liver injury, chronic kidney disease, and SCI, are better able to promote functional recovery in mice after SCI than extracellular vesicles derived from MSCs without preconditioning (EVs). MEVs were found to facilitate motor behavioral recovery more than EVs and to increase microglia/macrophages polarization from M1-like to M2-like in mice. Experiments in BV2 microglia and RAW264.7 macrophages confirmed that MEVs facilitate M2-like polarization and also showed that they reduce the production of reactive oxygen species (ROS) and regulate mitochondrial function. Proteomics analysis revealed that ubiquitin-specific protease 29 (USP29) was markedly increased in MEVs, and knockdown of USP29 in MEVs (shUSP29-MEVs) abolished MEVs-mediated benefits in vitro and in vivo. We then showed that USP29 interacts with, deubiquitinates and therefore stabilizes nuclear factor-like 2 (NRF2), thereby regulating microglia/macrophages polarization. In NRF2 knockout mice, MEVs failed to promote functional recovery and M2-like microglia/macrophages polarization. We also showed that MT reduced global N6-methyladenosine (m6 A) modification and levels of the m6 A "writer" methyltransferase-like 3 (METTL3). The stability of USP29 mRNA in MSCs was enhanced by treatment with MT, but inhibited by overexpression of METTL3. This study describes a very promising extracellular vesicle-based approach for treating SCI.
Collapse
Affiliation(s)
- Wei Liu
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Pengyu Tang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jiaxing Wang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Wu Ye
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xuhui Ge
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yuluo Rong
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chengyue Ji
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhuanghui Wang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jianling Bai
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jin Fan
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Guoyong Yin
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Weihua Cai
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
18
|
Ji R, Zhang X. The Roles of RNA N6-Methyladenosine in Regulating Stem Cell Fate. Front Cell Dev Biol 2021; 9:765635. [PMID: 34805173 PMCID: PMC8602194 DOI: 10.3389/fcell.2021.765635] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 09/20/2021] [Indexed: 01/12/2023] Open
Abstract
RNA N6-methyladenosine (m6A) modification has important regulatory roles in determining cell fate. The reversible methylation process of adding and removing m6A marks is dynamically regulated by a fine-tuned coordination of many enzymes and binding proteins. Stem cells have self-renewal and pluripotent potential and show broad prospects in regenerative medicine and other fields. Stem cells have also been identified in cancer, which is linked to cancer metastasis, therapy resistance, and recurrence. Herein, we aimed to review the molecular mechanism that controls the reversible balance of m6A level in stem cells and the effect of m6A modification on the balance between pluripotency and differentiation. Additionally, we also elaborated the association between aberrant m6A modification and the maintenance of cancer stem cells in many cancers. Moreover, we discussed about the clinical implications of m6A modification in cancer stem cells for cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Runbi Ji
- The Affiliated People's Hospital of Jiangsu University, Zhenjiang, China.,Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Xu Zhang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| |
Collapse
|
19
|
Liu Z, Cai Y, Deng M, Li D, Leng Q, Shi L, Tang Y, Wang F, Wan Y. Expression pattern of alkB homolog 5 in goat testis and its role in spermatogonial stem cells. Cell Tissue Res 2021; 387:131-142. [PMID: 34725717 DOI: 10.1007/s00441-021-03550-4] [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: 07/29/2021] [Accepted: 10/25/2021] [Indexed: 10/19/2022]
Abstract
RNA N6-methyladenosine (m6A) is essential for many bioprocesses in many species, but its role in goat testis development remains elusive, especially alkB homolog 5 (ALKBH5), one of the m6A demethylases. To this end, nine healthy Haimen goats of different ages were chosen randomly to provide testes. The results showed that the expression level of ALKBH5 was increased significantly (P < 0.05) in the 9-month group compared with the 0-day and 3-month groups, and ALKBH5 was located in goat spermatocytes with the highest expression level compared with Leydig cells and Sertoli cells. Thus, pcDNA3.1-ALKBH5 was constructed to explore the influences of the ALKBH5 increase in goat spermatogonial stem cells (SSC) in vitro. The results showed that the expression level of ALKBH5 in SSC transfected with pcDNA3.1-ALKBH5 (OE_ALKBH5) was significantly increased (P < 0.001) compared with that in SSC transfected with pcDNA3.1-EGFP (EGFP). With ALKBH5 overexpression in SSC, flow cytometry analysis showed that cells at G1 phase were significantly reduced (P < 0.01), while cells at S phase significantly increased (P < 0.01), and cell apoptosis was inhibited. Accordingly, the mRNA degradation of CCND1, CCNE1, and BCL2 was suppressed with ALKBH5 overexpression in SSC after treatment with actinomycin D. Furthermore, the mRNA levels of pluripotency maintenance- and cell differentiation-associated genes were changed between the two groups. Overall, the results indicated the crucial role of ALKBH5 during Haimen goat testis development. The results of this study provide a theoretical basis and technical means for RNA methylation participating in goat testis development.
Collapse
Affiliation(s)
- Zifei Liu
- Jiangsu Livestock Embryo Engineering Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, No. 1, Weigang, Nanjing, Jiangsu, 210095, China
| | - Yu Cai
- Jiangsu Livestock Embryo Engineering Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, No. 1, Weigang, Nanjing, Jiangsu, 210095, China
| | - Mingtian Deng
- Jiangsu Livestock Embryo Engineering Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, No. 1, Weigang, Nanjing, Jiangsu, 210095, China
| | - Dongxu Li
- Jiangsu Livestock Embryo Engineering Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, No. 1, Weigang, Nanjing, Jiangsu, 210095, China
| | - Qingqing Leng
- Jiangsu Livestock Embryo Engineering Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, No. 1, Weigang, Nanjing, Jiangsu, 210095, China
| | - Liangyue Shi
- Jiangsu Livestock Embryo Engineering Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, No. 1, Weigang, Nanjing, Jiangsu, 210095, China
| | - Yutong Tang
- Jiangsu Livestock Embryo Engineering Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, No. 1, Weigang, Nanjing, Jiangsu, 210095, China
| | - Feng Wang
- Jiangsu Livestock Embryo Engineering Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, No. 1, Weigang, Nanjing, Jiangsu, 210095, China
| | - Yongjie Wan
- Jiangsu Livestock Embryo Engineering Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, No. 1, Weigang, Nanjing, Jiangsu, 210095, China.
| |
Collapse
|
20
|
Si C, Chen C, Guo Y, Kang Q, Sun Z. Effect, Mechanism, and Applications of Coding/Non-coding RNA m6A Modification in Tumor Microenvironment. Front Cell Dev Biol 2021; 9:711815. [PMID: 34660577 PMCID: PMC8514707 DOI: 10.3389/fcell.2021.711815] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 08/30/2021] [Indexed: 01/07/2023] Open
Abstract
The tumor microenvironment (TME), which includes immune cells, fibroblasts, and other components, is the site of tumor cell growth and metastasis and significantly impacts tumor development. Among them, N6-methyladenosine RNA modifications (m6A RNA modifications) are the most abundant internal modifications in coding and non-coding RNAs, which can significantly influence the cancer process and have potential as biomarkers and potential therapeutic targets for tumor therapy. This manuscript reviews the role of m6A RNA modifications in TME and their application in tumor therapy. To some extent, an in-depth understanding of the relationship between TME and m6A RNA modifications will provide new approaches and ideas for future cancer therapy.
Collapse
Affiliation(s)
- Chaohua Si
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,School of Life Sciences, Zhengzhou University, Zhengzhou, China
| | - Chen Chen
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| | - Yaxin Guo
- Henan Academy of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China.,School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Qiaozhen Kang
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| | - Zhenqiang Sun
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
21
|
Loh D, Reiter RJ. Melatonin: Regulation of Biomolecular Condensates in Neurodegenerative Disorders. Antioxidants (Basel) 2021; 10:1483. [PMID: 34573116 PMCID: PMC8465482 DOI: 10.3390/antiox10091483] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/10/2021] [Accepted: 09/13/2021] [Indexed: 12/12/2022] Open
Abstract
Biomolecular condensates are membraneless organelles (MLOs) that form dynamic, chemically distinct subcellular compartments organizing macromolecules such as proteins, RNA, and DNA in unicellular prokaryotic bacteria and complex eukaryotic cells. Separated from surrounding environments, MLOs in the nucleoplasm, cytoplasm, and mitochondria assemble by liquid-liquid phase separation (LLPS) into transient, non-static, liquid-like droplets that regulate essential molecular functions. LLPS is primarily controlled by post-translational modifications (PTMs) that fine-tune the balance between attractive and repulsive charge states and/or binding motifs of proteins. Aberrant phase separation due to dysregulated membrane lipid rafts and/or PTMs, as well as the absence of adequate hydrotropic small molecules such as ATP, or the presence of specific RNA proteins can cause pathological protein aggregation in neurodegenerative disorders. Melatonin may exert a dominant influence over phase separation in biomolecular condensates by optimizing membrane and MLO interdependent reactions through stabilizing lipid raft domains, reducing line tension, and maintaining negative membrane curvature and fluidity. As a potent antioxidant, melatonin protects cardiolipin and other membrane lipids from peroxidation cascades, supporting protein trafficking, signaling, ion channel activities, and ATPase functionality during condensate coacervation or dissolution. Melatonin may even control condensate LLPS through PTM and balance mRNA- and RNA-binding protein composition by regulating N6-methyladenosine (m6A) modifications. There is currently a lack of pharmaceuticals targeting neurodegenerative disorders via the regulation of phase separation. The potential of melatonin in the modulation of biomolecular condensate in the attenuation of aberrant condensate aggregation in neurodegenerative disorders is discussed in this review.
Collapse
Affiliation(s)
- Doris Loh
- Independent Researcher, Marble Falls, TX 78654, USA
| | - Russel J. Reiter
- Department of Cellular and Structural Biology, UT Health Science Center, San Antonio, TX 78229, USA
| |
Collapse
|
22
|
METTL3 Intensifies the Progress of Oral Squamous Cell Carcinoma via Modulating the m6A Amount of PRMT5 and PD-L1. J Immunol Res 2021; 2021:6149558. [PMID: 34476262 PMCID: PMC8408004 DOI: 10.1155/2021/6149558] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 07/26/2021] [Accepted: 08/02/2021] [Indexed: 12/19/2022] Open
Abstract
N6-Methyladenosine (m6A) modification is one of the commonest chemical modifications in eukaryotic mRNAs, which has essential effects on mRNA translation, splicing, and stability. Currently, there is a rising concern on the regulatory role of m6A in tumorigenesis. As a known component in the m6A methyltransferase complex, METTL3 (methyltransferase-like 3) plays an essential role in m6A methylation. Till now, the functions of METTL3 in oral squamous cell carcinoma (OSCC) and its relative mechanism remain to be explored. In this research, through the GEPIA database, we found that high METTL3 expression has a correlation with poor prognosis of squamous cell carcinoma of head and neck. qRT-PCR displayed that METTL3 was highly expressed in OSCC cells. Functionally, METTL3 knockdown reduced the invasion, migration, and proliferation competence of OSCC cells and attenuated the activation of CD8+ T cells. In contrast, METTL3 overexpression resulted in opposite results. GEPIA, UALCAN, and SRAMP databases, PCR, western blot, and m6A RNA methylation assay confirmed the m6A modification of PRMT5 and PD-L1 mediated by METTL3. In conclusion, our results displayed that METTL3 intensified the metastasis and proliferation of OSCC by modulating the m6A amounts of PRMT5 and PD-L1, suggesting that METTL3 may be a therapeutic target for OSCC patients.
Collapse
|
23
|
Guo W, Tan F, Huai Q, Wang Z, Shao F, Zhang G, Yang Z, Li R, Xue Q, Gao S, He J. Comprehensive Analysis of PD-L1 Expression, Immune Infiltrates, and m6A RNA Methylation Regulators in Esophageal Squamous Cell Carcinoma. Front Immunol 2021; 12:669750. [PMID: 34054840 PMCID: PMC8149800 DOI: 10.3389/fimmu.2021.669750] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 04/28/2021] [Indexed: 12/21/2022] Open
Abstract
Background Esophageal squamous cell carcinoma (ESCC) is one of the most common cancer types and represents a threat to global public health. N6-Methyladenosine (m6A) methylation plays a key role in the occurrence and development of many tumors, but there are still few studies investigating ESCC. This study attempts to construct a prognostic signature of ESCC based on m6A RNA methylation regulators and to explore the potential association of these regulators with the tumor immune microenvironment (TIME). Methods The transcriptome sequencing data and clinical information of 20 m6A RNA methylation regulators in 453 patients with ESCC (The Cancer Genome Atlas [TCGA] cohort, n = 95; Gene Expression Omnibus [GEO] cohort, n = 358) were obtained. The differing expression levels of m6A regulators between ESCC and normal tissue were evaluated. Based on the expression of these regulators, consensus clustering was performed to investigate different ESCC clusters. PD-L1 expression, immune score, immune cell infiltration and potential mechanisms among different clusters were examined. LASSO Cox regression analysis was utilized to obtain a prognostic signature based on m6A RNA methylation modulators. The relationship between the risk score based on the prognostic signature and the TIME of ESCC patients was studied in detail. Results Six m6A regulators (METTL3, WTAP, IGF2BP3, YTHDF1, HNRNPA2B1 and HNRNPC) were observed to be significantly highly expressed in ESCC tissues. Two molecular subtypes (clusters 1/2) were determined by consensus clustering of 20 m6A modulators. The expression level of PD-L1 in ESCC tissues increased significantly and was significantly negatively correlated with the expression levels of YTHDF2, METL14 and KIAA1429. The immune score, CD8 T cells, resting mast cells, and regulatory T cells (Tregs) in cluster 2 were significantly increased. Gene set enrichment analysis (GSEA) shows that this cluster involves multiple hallmark pathways. We constructed a five-gene prognostic signature based on m6A RNA methylation, and the risk score based on the prognostic signature was determined to be an independent prognostic indicator of ESCC. More importantly, the prognostic value of the prognostic signature was verified using another independent cohort. m6A regulators are related to TIME, and their copy-number alterations will dynamically affect the number of tumor-infiltrating immune cells. Conclusion Our study established a strong prognostic signature based on m6A RNA methylation regulators; this signature was able to accurately predict the prognosis of ESCC patients. The m6A methylation regulator may be a key mediator of PD-L1 expression and immune cell infiltration and may strongly affect the TIME of ESCC.
Collapse
Affiliation(s)
- Wei Guo
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fengwei Tan
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qilin Huai
- Department of Graduate School, Zunyi Medical University, Zunyi, China
| | - Zhen Wang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fei Shao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Cancer Institute of The Affiliated Hospital of Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong, China
| | - Guochao Zhang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhenlin Yang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Renda Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qi Xue
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shugeng Gao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
24
|
He X, Zhu Y, Yang L, Wang Z, Wang Z, Feng J, Wen X, Cheng L, Zhu R. MgFe-LDH Nanoparticles: A Promising Leukemia Inhibitory Factor Replacement for Self-Renewal and Pluripotency Maintenance in Cultured Mouse Embryonic Stem Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2003535. [PMID: 33977050 PMCID: PMC8097378 DOI: 10.1002/advs.202003535] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 01/15/2021] [Indexed: 05/20/2023]
Abstract
Leukemia inhibitory factor (LIF), an indispensable bioactive protein that sustains self-renewal and pluripotency in stem cells, is vital for mouse embryonic stem cell (mESC) culture. Extensive research is conducted on reliable alternatives for LIF as its clinical application in stable culture and large-scale expansion of ESCs is limited by its instability and high cost. However, few studies have sought to replace LIF with nanoparticles to provide a xeno-free culture condition. MgAl-LDH (layered double hydroxide) nanoparticles can partially replace LIF in maintaining pluripotency of mESCs; however, the requirement and tolerance for aluminum ions in mice are far lesser than those of iron ions. Hence, MgFe-LDH nanoparticles are selected for this study. MgFe-LDH is superior to MgAl-LDH in maintaining self-renewal and pluripotency of mESCs, in the absence of LIF and mouse embryonic fibroblast. Furthermore, combined transcriptomic and proteomic analysis confirms that MgFe-LDH can activate the LIF receptor (LIFR)/phosphatidylinositol 3-kinase (PI3K)/protein kinase B(AKT), LIFR/JAK/janus kinase (JAK)/signal transducer and activator of transcription 3(STAT3), and phospho-signal transducer and activator of transcription 3(p-STAT3)/ten-eleven translocation (TET) signaling pathways, while the extra Fe2+ provided by MgFe-LDH would also enhance TET1/2 abundance thus affecting the TET1/2 regulated pluripotency related marker expression and TET1/2 meditated DNA demethylation. These results suggest that MgFe-LDH nanoparticles can thus be used as an affordable and efficient replacement for LIF in mESC cultivation.
Collapse
Affiliation(s)
- Xiaolie He
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of EducationOrthopaedic Department of Tongji HospitalSchool of Life Science and TechnologyTongji University389 Xincun RoadShanghai200065P. R. China
| | - Yanjing Zhu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of EducationOrthopaedic Department of Tongji HospitalSchool of Life Science and TechnologyTongji University389 Xincun RoadShanghai200065P. R. China
| | - Li Yang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of EducationOrthopaedic Department of Tongji HospitalSchool of Life Science and TechnologyTongji University389 Xincun RoadShanghai200065P. R. China
| | - Zhaojie Wang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of EducationOrthopaedic Department of Tongji HospitalSchool of Life Science and TechnologyTongji University389 Xincun RoadShanghai200065P. R. China
| | - Zekun Wang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of EducationOrthopaedic Department of Tongji HospitalSchool of Life Science and TechnologyTongji University389 Xincun RoadShanghai200065P. R. China
| | - Jianhao Feng
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of EducationOrthopaedic Department of Tongji HospitalSchool of Life Science and TechnologyTongji University389 Xincun RoadShanghai200065P. R. China
| | - Xuejun Wen
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of EducationOrthopaedic Department of Tongji HospitalSchool of Life Science and TechnologyTongji University389 Xincun RoadShanghai200065P. R. China
- Department of Chemical and Life Science EngineeringSchool of EngineeringVirginia Commonwealth UniversityRichmondVA23284USA
| | - Liming Cheng
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of EducationOrthopaedic Department of Tongji HospitalSchool of Life Science and TechnologyTongji University389 Xincun RoadShanghai200065P. R. China
| | - Rongrong Zhu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of EducationOrthopaedic Department of Tongji HospitalSchool of Life Science and TechnologyTongji University389 Xincun RoadShanghai200065P. R. China
| |
Collapse
|
25
|
Dai DL, Li X, Wang L, Xie C, Jin Y, Zeng MS, Zuo Z, Xia TL. Identification of an N6-methyladenosine-mediated positive feedback loop that promotes Epstein-Barr virus infection. J Biol Chem 2021; 296:100547. [PMID: 33741341 PMCID: PMC8063736 DOI: 10.1016/j.jbc.2021.100547] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 03/10/2021] [Accepted: 03/15/2021] [Indexed: 12/11/2022] Open
Abstract
N6-methyladenosine (m6A) is among the most abundant mRNA modifications, particularly in eukaryotes, and is found in mammals, plants, and even some viruses. Although essential for the regulation of many biological processes, the exact role of m6A modification in virus–host interaction remains largely unknown. Here, using m6A -immunoprecipitation and sequencing, we find that Epstein–Barr virus (EBV) infection decreases the m6A modification of transcriptional factor KLF4 mRNA and subsequently increases its protein level. Mechanistically, EBV immediate-early protein BZLF1 interacts with the promoter of m6A methyltransferase METTL3, inhibiting its expression. Subsequently, the decrease of METTL3 reduces the level of KLF4 mRNA m6A modification, preventing its decay by the m6A reader protein YTHDF2. As a result, KLF4 protein level is upregulated and, in turn, promotes EBV infection of nasopharyngeal epithelial cells. Thus, our results suggest the existence of a positive feedback loop formed between EBV and host molecules via cellular mRNA m6A levels, and this feedback loop acts to facilitate viral infection. This mechanism contains multiple potential targets for controlling viral infectious diseases.
Collapse
Affiliation(s)
- Dan-Ling Dai
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Xingyang Li
- Department of Temporomandibular Joint Surgery, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, P. R. China
| | - Lin Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China; Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Chu Xie
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Yanan Jin
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China; Department of VIP Region, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Mu-Sheng Zeng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Zhixiang Zuo
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China.
| | - Tian-Liang Xia
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China.
| |
Collapse
|
26
|
Zhang L, Wang X, Zhang H, Feng M, Ding J, Zhang B, Cheng Z, Qian L. Exercise-induced peptide EIP-22 protect myocardial from ischaemia/reperfusion injury via activating JAK2/STAT3 signalling pathway. J Cell Mol Med 2021; 25:3560-3572. [PMID: 33710777 PMCID: PMC8034444 DOI: 10.1111/jcmm.16441] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 02/20/2021] [Accepted: 02/26/2021] [Indexed: 02/06/2023] Open
Abstract
Recent studies have revealed that exercise has myocardial protective effects, but the exact mechanism remains unclear. Studies have increasingly found that peptides play a protective role in myocardial ischaemia‐reperfusion (I/R) injury. However, little is known about the role of exercise‐induced peptides in myocardial I/R injury. To elucidate the effect of exercise‐induced peptide EIP‐22 in myocardial I/R injury, we first determined the effect of EIP‐22 on hypoxia/reperfusion (H/R)‐ or H2O2‐induced injury via assessing cell viability and lactate dehydrogenase (LDH) level. In addition, reactive oxygen species (ROS) accumulation and mitochondrial membrane potential (MMP) was assessed by fluorescence microscope. Meanwhile, Western blot and TUNEL methods were used to detect apoptosis level. Then, we conducted mice I/R injury model and verified the effect of EIP‐22 by measuring cardiac function, evaluating heart pathology and detecting serum LDH, CK‐MB and cTnI level. Finally, the main signalling pathway was analysed by RNA‐seq. In vitro, EIP‐22 treatment significantly improved cells viabilities and MMP and attenuated the LDH, ROS and apoptosis level. In vivo, EIP‐22 distinctly improved cardiac function, ameliorated myocardial infarction area and fibrosis and decreased serum LDH, CK‐MB and cTnI level. Mechanistically, JAK/STAT signalling pathway was focussed by RNA‐seq and we confirmed that EIP‐22 up‐regulated the expression of p‐JAK2 and p‐STAT3. Moreover, AG490, a selective inhibitor of JAK2/STAT3, eliminated the protective roles of EIP‐22. The results uncovered that exercise‐induced peptide EIP‐22 protected cardiomyocytes from myocardial I/R injury via activating JAK2/STAT3 signalling pathway and might be a new candidate molecule for the treatment of myocardial I/R injury.
Collapse
Affiliation(s)
- Li Zhang
- Department of Cardiology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xuejun Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hao Zhang
- Department of Internal Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Mengwen Feng
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jingjing Ding
- Department of General Practice, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bing Zhang
- Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zijie Cheng
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Lingmei Qian
- Department of Cardiology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
27
|
Guo W, Huai Q, Wan H, Guo L, Song P, Gao S, He J. Prognostic Impact of IGF2BP3 Expression in Patients with Surgically Resected Lung Adenocarcinoma. DNA Cell Biol 2021; 40:316-331. [PMID: 33493403 DOI: 10.1089/dna.2020.6136] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Lung adenocarcinoma (LUAD) acts as the representative histological subtype regarding lung cancer, and its mortality and morbidity remain high. Therefore, more therapeutic targets are needed. Insulin-like growth factor 2 messenger RNA-binding proteins (IGF2BPs), a class of RNA-binding proteins, including IGF2BP1, IGF2BP2, and IGF2BP3, are considered to be the "reader" of N6-methyladenosine (m6A) methylation and remarkably affects cancer occurrence and development. Studies have shown that IGF2BP3 has prognostic potential in multiple public databases compared with other members of the IGF2BPs family. This research aims to use the expression data of IGF2BP3 in multiple cohorts to explore the expression as well as prognostic significance of IGF2BP3 in LUAD. Based on the six datasets from the Oncomine database, LUAD tissue presented obviously higher IGF2BP3 expression relative to normal tissue. In the three cohorts of The Cancer Genome Atlas (TCGA), Gene expression profiling interactive analysis, and Gene Expression Omnibus, IGF2BP3 was abnormally highly expressed in LUAD, and it could lead to a weaker prognosis (p < 0.05). The cohort from National Cancer Center of China (NCC) verified the prognostic value of IGF2BP3, and the high IGF2BP3 expression could be remarkably related to gender, tumor length, differentiation, and T stage (p < 0.05). Cox regression analysis of TCGA and NCC cohorts consistently showed that IGF2BP3 expression could serve for independently predicting the prognosis of LUAD patients (p < 0.05). Ten hallmark pathways with significant enrichment were identified. IGF2BP3 is abnormally highly expressed in LUAD tissue, and can lead to worse overall survival. In summary, IGF2BP3 may be an oncogene and potential prognostic biomarker of LUAD.
Collapse
Affiliation(s)
- Wei Guo
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Qilin Huai
- Department of Graduate School, Zunyi Medical University, Zunyi, People's Republic of China
| | - Huan Wan
- PET-CT Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Lei Guo
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Peng Song
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Shugeng Gao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| |
Collapse
|