1
|
Han X, Zhu Y, Ke J, Zhai Y, Huang M, Zhang X, He H, Zhang X, Zhao X, Guo K, Li X, Han Z, Zhang Y. Progression of m 6A in the tumor microenvironment: hypoxia, immune and metabolic reprogramming. Cell Death Discov 2024; 10:331. [PMID: 39033180 PMCID: PMC11271487 DOI: 10.1038/s41420-024-02092-2] [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: 12/24/2023] [Revised: 07/03/2024] [Accepted: 07/05/2024] [Indexed: 07/23/2024] Open
Abstract
Recently, N6-methyladenosine (m6A) has aroused widespread discussion in the scientific community as a mode of RNA modification. m6A comprises writers, erasers, and readers, which regulates RNA production, nuclear export, and translation and is very important for human health. A large number of studies have found that the regulation of m6A is closely related to the occurrence and invasion of tumors, while the homeostasis and function of the tumor microenvironment (TME) determine the occurrence and development of tumors to some extent. TME is composed of a variety of immune cells (T cells, B cells, etc.) and nonimmune cells (tumor-associated mesenchymal stem cells (TA-MSCs), cancer-associated fibroblasts (CAFs), etc.). Current studies suggest that m6A is involved in regulating the function of various cells in the TME, thereby affecting tumor progression. In this manuscript, we present the composition of m6A and TME, the relationship between m6A methylation and characteristic changes in TME, the role of m6A methylation in TME, and potential therapeutic strategies to provide new perspectives for better treatment of tumors in clinical work.
Collapse
Affiliation(s)
- Xuan Han
- First Clinical College of Changzhi Medical College, Changzhi, China
| | - Yu Zhu
- Linfen Central Hospital, Linfen, China
| | - Juan Ke
- Linfen Central Hospital, Linfen, China
| | | | - Min Huang
- Linfen Central Hospital, Linfen, China
| | - Xin Zhang
- Linfen Central Hospital, Linfen, China
| | | | | | | | | | | | - Zhongyu Han
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | | |
Collapse
|
2
|
Chen Y, Liu M, Lu M, Luo L, Han Z, Liu X. Exploring the impact of m 6A modification on immune diseases: mechanisms and therapeutic implication. Front Immunol 2024; 15:1387582. [PMID: 39072324 PMCID: PMC11272477 DOI: 10.3389/fimmu.2024.1387582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 06/28/2024] [Indexed: 07/30/2024] Open
Abstract
N6-methyladenosine (m6A) is a chemical modification of RNA and has become a widely discussed topic among scientific researchers in recent years. It is distributed in various organisms, including eukaryotes and bacteria. It has been found that m6A is composed of writers, erasers and readers and is involved in biological functions such as splicing, transport and translation of RNA. The balance of the human immune microenvironment is important for human health abnormalities. Increasing studies have found that m6A affects the development of immune diseases such as inflammatory enteritis and systemic lupus erythematosus (SLE) by participating in the homeostatic regulation of the immune microenvironment in vivo. In this manuscript, we introduce the composition, biological function, regulation of m6A in the immune microenvironment and its progression in various immune diseases, providing new targets and directions for the treatment of immune diseases in clinical practice.
Collapse
Affiliation(s)
- Yutong Chen
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Min Liu
- Department of Traditional Chinese Medicine, Zhejiang Hospital of Integrated Traditional Chinese and Western Medicine, Hangzhou, Zhejiang, China
| | - Miao Lu
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Linling Luo
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhongyu Han
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xide Liu
- Department of Traditional Chinese Medicine, Zhejiang Hospital of Integrated Traditional Chinese and Western Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
3
|
Mashayekhi V, Schomisch A, Rasheed S, Aparicio-Puerta E, Risch T, Yildiz D, Koch M, Both S, Ludwig N, Legroux TM, Keller A, Müller R, Fuhrmann G, Hoppstädter J, Kiemer AK. The RNA binding protein IGF2BP2/IMP2 alters the cargo of cancer cell-derived extracellular vesicles supporting tumor-associated macrophages. Cell Commun Signal 2024; 22:344. [PMID: 38937789 PMCID: PMC11212187 DOI: 10.1186/s12964-024-01701-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 06/05/2024] [Indexed: 06/29/2024] Open
Abstract
BACKGROUND Tumor cells release extracellular vesicles (EVs) that contribute to the polarization of macrophages towards tumor-associated macrophages (TAMs). High expression levels of the RNA binding protein IGF2BP2/IMP2 are correlated with increased tumor cell proliferation, invasion, and poor prognosis in the clinic. However, there is a lack of understanding of whether IMP2 affects the cargo of cancer cell-derived EVs, thereby modulating macrophage polarization. METHODS EVs were isolated from IMP2-expressing HCT116 parental cells (WT) and CRISPR/Cas9 IMP2 knockout (KO) cells. EVs were characterized according to MISEV guidelines, microRNA cargo was assessed by microRNA-Seq, and the protein cargo was analyzed by proteomics. Primary human monocyte-derived macrophages (HMDMs) were polarized by EVs, and the expression of genes and surface markers was assessed using qPCR and flow cytometry, respectively. Morphological changes of macrophages, as well as the migratory potential of cancer cells, were assessed by the Incucyte® system and macrophage matrix degradation potential by zymography. Changes in the metabolic activity of macrophages were quantified using a Seahorse® analyzer. For in vivo studies, EVs were injected into the yolk sac of zebrafish larvae, and macrophages were isolated by fluorescence-activated cell sorting. RESULTS EVs from WT and KO cells had a similar size and concentration and were positive for 25 vesicle markers. The expression of tumor-promoting genes was higher in macrophages polarized with WT EVs than KO EVs, while the expression of TNF and IL6 was reduced. A similar pattern was observed in macrophages from zebrafish larvae treated in vivo. WT EV-polarized macrophages showed a higher abundance of TAM-like surface markers, higher matrix degrading activity, as well as a higher promotion of cancer cell migration. MicroRNA-Seq revealed a significant difference in the microRNA composition of WT and KO EVs, particularly a high abundance of miR-181a-5p in WT EVs, which was absent in KO EVs. Inhibitors of macropinocytosis and phagocytosis antagonized the delivery of miR-181a-5p into macrophages and the downregulation of the miR-181a-5p target DUSP6. Proteomics data showed differences in protein cargo in KO vs. WT EVs, with the differentially abundant proteins mainly involved in metabolic pathways. WT EV-treated macrophages exhibited a higher basal oxygen consumption rate and a lower extracellular acidification rate than KO EV-treated cells. CONCLUSION Our results show that IMP2 determines the cargo of EVs released by cancer cells, thereby modulating the EVs' actions on macrophages. Expression of IMP2 is linked to the secretion of EVs that polarize macrophages towards a tumor-promoting phenotype.
Collapse
Affiliation(s)
- Vida Mashayekhi
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, 66123, Saarbrücken, Germany
| | - Annika Schomisch
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, 66123, Saarbrücken, Germany
| | - Sari Rasheed
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), and Department of Pharmacy, Saarland University, Saarbrücken, Germany
- German Centre for Infection Research (DZIF), Brunswick, Germany
| | - Ernesto Aparicio-Puerta
- Chair for Clinical Bioinformatics, Saarland University, University Hospital, Saarbrücken, Germany
| | - Timo Risch
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), and Department of Pharmacy, Saarland University, Saarbrücken, Germany
- German Centre for Infection Research (DZIF), Brunswick, Germany
| | - Daniela Yildiz
- Institute of Experimental and Clinical Pharmacology and Toxicology, PZMS, ZHMB, Saarland University, Homburg, Germany
| | - Marcus Koch
- INM - Leibniz Institute for New Materials, Saarbrücken, Germany
| | - Simon Both
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, 66123, Saarbrücken, Germany
| | - Nicole Ludwig
- Department of Human Genetics, Saarland University, Homburg, Germany
| | - Thierry M Legroux
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, 66123, Saarbrücken, Germany
| | - Andreas Keller
- Chair for Clinical Bioinformatics, Saarland University, University Hospital, Saarbrücken, Germany
| | - Rolf Müller
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), and Department of Pharmacy, Saarland University, Saarbrücken, Germany
- German Centre for Infection Research (DZIF), Brunswick, Germany
- Department of Pharmacy, Saarland University, Saarbrücken, Germany
| | - Gregor Fuhrmann
- Department of Pharmaceutical Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Jessica Hoppstädter
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, 66123, Saarbrücken, Germany
| | - Alexandra K Kiemer
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, 66123, Saarbrücken, Germany.
| |
Collapse
|
4
|
Nan Y, Chen M, Wu W, Huang R, Sun W, Lu Q, Gu Z, Mao X, Xu H, Wang Y. IGF2BP2 regulates the inflammation of fibroblast-like synoviocytes via GSTM5 in rheumatoid arthritis. Cell Death Discov 2024; 10:215. [PMID: 38702323 PMCID: PMC11068746 DOI: 10.1038/s41420-024-01988-3] [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: 08/05/2023] [Revised: 04/19/2024] [Accepted: 04/23/2024] [Indexed: 05/06/2024] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease with an unknown etiology. RA cannot be fully cured and requires lengthy treatment, imposing a significant burden on both individuals and society. Due to the lack of specific drugs available for treating RA, exploring a key new therapeutic target for RA is currently an important task. Activated fibroblast-like synoviocytes (FLSs) play a crucial role in the progression of RA, which release interleukin (IL)-1β, IL-6 and tumor necrosis factor (TNF)-α resulting in abnormal inflammatory reaction in the synovium. A previous study has highlighted the correlation of m6A reader insulin-like growth factor 2 mRNA-binding protein 2 (IGF2BP2) with inflammation-related diseases in human. However, the role of IGF2BP2 in the inflammatory reaction of FLSs during RA progression has not been assessed. In this study, IGF2BP2 expression was decreased in the synovial tissues of RA patients and collagen-induced arthritis (CIA) rats. Intra-articular injection of an adeno-associated virus (AAV) vector overexpressing IGF2BP2 relieved paw swelling, synovial hyperplasia and cartilage destruction in CIA rats. IGF2BP2 overexpression also inhibited lipopolysaccharide (LPS)-mediated RA fibroblast-like synoviocytes (RA-FLSs) migration and invasion accompanied by a decreased level of inflammatory factors in vitro. Conversely, IGF2BP2 suppression promoted RA-FLSs migration and invasion with an elevated level of inflammatory factors in vitro. The sequencing result showed that glutathione S-transferase Mu 5 (GSTM5), a key antioxidant gene, was the target mRNA of IGF2BP2. Further experiments demonstrated that IGF2BP2 strengthened the stability of GSTM5 mRNA, leading to weakened inflammatory reaction and reduced expression of matrix metalloproteinase 9 and 13 (MMP9, MMP13). Therefore, IGF2BP2-GSTM5 axis may represent a potential therapeutic target for RA treatment.
Collapse
Affiliation(s)
- Yunyi Nan
- Department of Orthopaedics, Affiliated Hospital of Nantong University, Medical School of Nantong University, 226001, Nantong, China
| | - Minhao Chen
- Department of Orthopaedics, Affiliated Hospital of Nantong University, Medical School of Nantong University, 226001, Nantong, China
| | - Weijie Wu
- Department of Orthopaedics, Affiliated Hospital of Nantong University, Medical School of Nantong University, 226001, Nantong, China
- Department of Orthopaedics, Affiliated Nantong Hospital of Shanghai University, The Sixth People's Hospital of Nantong, 226001, Nantong, China
| | - Rongrong Huang
- Department of Pharmacy, Affiliated Hospital of Nantong University, 226001, Nantong, China
| | - Weiwei Sun
- Department of Orthopaedics, Affiliated Hospital of Nantong University, Medical School of Nantong University, 226001, Nantong, China
| | - Qian Lu
- Department of Rheumatology, Affiliated Hospital of Nantong University, 226001, Nantong, China
| | - Zhifeng Gu
- Department of Rheumatology, Affiliated Hospital of Nantong University, 226001, Nantong, China
| | - Xingxing Mao
- Department of Orthopaedics, Affiliated Nantong Hospital of Shanghai University, The Sixth People's Hospital of Nantong, 226001, Nantong, China.
| | - Hua Xu
- Department of Orthopaedics, Affiliated Hospital of Nantong University, Medical School of Nantong University, 226001, Nantong, China.
| | - Youhua Wang
- Department of Orthopaedics, Affiliated Hospital of Nantong University, Medical School of Nantong University, 226001, Nantong, China.
| |
Collapse
|
5
|
Chen S, Saeed AFUH, Liu Q, Jiang Q, Xu H, Xiao GG, Rao L, Duo Y. Macrophages in immunoregulation and therapeutics. Signal Transduct Target Ther 2023; 8:207. [PMID: 37211559 DOI: 10.1038/s41392-023-01452-1] [Citation(s) in RCA: 312] [Impact Index Per Article: 312.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 03/06/2023] [Accepted: 04/26/2023] [Indexed: 05/23/2023] Open
Abstract
Macrophages exist in various tissues, several body cavities, and around mucosal surfaces and are a vital part of the innate immune system for host defense against many pathogens and cancers. Macrophages possess binary M1/M2 macrophage polarization settings, which perform a central role in an array of immune tasks via intrinsic signal cascades and, therefore, must be precisely regulated. Many crucial questions about macrophage signaling and immune modulation are yet to be uncovered. In addition, the clinical importance of tumor-associated macrophages is becoming more widely recognized as significant progress has been made in understanding their biology. Moreover, they are an integral part of the tumor microenvironment, playing a part in the regulation of a wide variety of processes including angiogenesis, extracellular matrix transformation, cancer cell proliferation, metastasis, immunosuppression, and resistance to chemotherapeutic and checkpoint blockade immunotherapies. Herein, we discuss immune regulation in macrophage polarization and signaling, mechanical stresses and modulation, metabolic signaling pathways, mitochondrial and transcriptional, and epigenetic regulation. Furthermore, we have broadly extended the understanding of macrophages in extracellular traps and the essential roles of autophagy and aging in regulating macrophage functions. Moreover, we discussed recent advances in macrophages-mediated immune regulation of autoimmune diseases and tumorigenesis. Lastly, we discussed targeted macrophage therapy to portray prospective targets for therapeutic strategies in health and diseases.
Collapse
Affiliation(s)
- Shanze Chen
- Department of Respiratory Diseases and Critic Care Unit, Shenzhen Institute of Respiratory Disease, Shenzhen Key Laboratory of Respiratory Disease, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, China
| | - Abdullah F U H Saeed
- Department of Cancer Biology, Beckman Research Institute of City of Hope National Medical Center, Los Angeles, CA, 91010, USA
| | - Quan Liu
- Department of Laboratory Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital), Shenzhen University, Shenzhen, 518052, China
| | - Qiong Jiang
- Department of Respiratory Diseases and Critic Care Unit, Shenzhen Institute of Respiratory Disease, Shenzhen Key Laboratory of Respiratory Disease, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, China
| | - Haizhao Xu
- Department of Respiratory Diseases and Critic Care Unit, Shenzhen Institute of Respiratory Disease, Shenzhen Key Laboratory of Respiratory Disease, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, China
- Department of Respiratory, The First Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Gary Guishan Xiao
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Sciences, School of Chemical Engineering, Dalian University of Technology, Dalian, China.
| | - Lang Rao
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, 518132, China.
| | - Yanhong Duo
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
6
|
m6A modification in inflammatory bowel disease provides new insights into clinical applications. Biomed Pharmacother 2023; 159:114298. [PMID: 36706633 DOI: 10.1016/j.biopha.2023.114298] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/13/2023] [Accepted: 01/20/2023] [Indexed: 01/26/2023] Open
Abstract
Inflammatory bowel disease (IBD) results from a complex interplay between genetic predisposition, environmental factors, and gut microbes. The role of N6-methyladenosine (m6A) methylation in the pathogenesis of IBD has attracted increasing attention. m6A modification not only regulates intestinal mucosal immunity and intestinal barrier function, but also affects apoptosis and autophagy in intestinal epithelial cells. Additionally, m6A modification participated in the interaction between gut microbes and the host, providing a novel direction to explore the molecular mechanisms of IBD and the theoretical basis for specific microorganism-oriented prevention and treatment measures. m6A regulators are expected to be biomarkers for predicting the prognosis of IBD patients. m6A methylation may be utilized as a novel target in the management of IBD. This review focused on the recent advances in how m6A modification causes the initiation and development of IBD, and provided new insights into optimal prevention and treatment measures for IBD.
Collapse
|
7
|
Wan L, Liu J, Huang C, Zhu Z, Li F, Sun G, Wang K, Li S, Ma X, Chen X, Yuan W. Role of m6A modification and novel circ_0066715/ miR-486-5p/ ETS1 axis in rheumatoid arthritis macrophage polarization progression. Aging (Albany NY) 2022; 14:10009-10026. [PMID: 36541909 PMCID: PMC9831719 DOI: 10.18632/aging.204439] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 12/09/2022] [Indexed: 01/06/2023]
Abstract
Rheumatoid arthritis (RA) is a systemic disease dominated by inflammatory synovitis. RA synovial macrophages tend undergo M1-type macrophage polarization. Then, polarized M1-type macrophages secrete abundant pro-inflammatory cytokines, causing joint and cartilage destruction. N6-methyladenosine (m6A) methylation modification, circular RNA (circRNA), microRNA (miRNA), messenger RNA (mRNA), etc. are involved in the inflammatory response of RA. We found that there is an imbalance of inflammatory polarization in RA, which is manifested by a sharp increase in inflammatory markers and a high inflammatory response. Here, we show that RA was closely associated with low expression of circ_0066715. The overexpression of circ_0066715 significantly increased the ETS1 levels in RA-FLS cells, decreased cytokine secretion by M1-type macrophages, elevated M2-type cytokines, and inhibited FLS proliferation. Interestingly, the overexpression of miR-486-5p significantly suppressed the attenuation of the cell function and the effect on M1 macrophage polarization caused by circ_0066715 positive expression. WTAP may be involved in the methylation process of ETS1 in RA. ETS1 m6A methylation levels were altered upon WTAP intervention. The overexpression or interference of circ_0066715 decreased or increased WTAP expression. Our findings provide a novel circRNA/miRNA/mRNA regulatory axis and m6A regulatory mechanism involved in the process of RA macrophage polarization, thereby providing a powerful diagnostic and therapeutic strategy for RA treatment.
Collapse
Affiliation(s)
- Lei Wan
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230038, China,Key Laboratory of Xin’an Medical Education Ministry, Hefei 230038, China
| | - Jian Liu
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230038, China,Key Laboratory of Xin’an Medical Education Ministry, Hefei 230038, China
| | - Chuanbing Huang
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230038, China
| | - Ziheng Zhu
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230038, China
| | - Fangze Li
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230038, China
| | - Guanghan Sun
- College of Traditional Chinese Medicine, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Kun Wang
- Key Laboratory of Xin’an Medical Education Ministry, Hefei 230038, China,College of Traditional Chinese Medicine, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Shu Li
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230038, China
| | - Ximeng Ma
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230038, China
| | - Xi Chen
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230038, China
| | - Wang Yuan
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230038, China
| |
Collapse
|
8
|
Wan L, Liu J, Huang C, Zhu Z, Wang K, Sun G, Zhu L, Hu Z. Comprehensive Analysis and Functional Characteristics of Differential Expression of N6-Methyladenosine Methylation Modification in the Whole Transcriptome of Rheumatoid Arthritis. Mediators Inflamm 2022; 2022:4766992. [PMID: 36330380 PMCID: PMC9626244 DOI: 10.1155/2022/4766992] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 09/22/2022] [Indexed: 08/04/2023] Open
Abstract
N6-methyladenosine (m6A) modification is the most prevalent chemical modification in eukaryotic mRNA and is associated with the development of various immune diseases. However, the role of m6A methylation in rheumatoid arthritis (RA) development is unclear. We preliminarily explored the role of m6A methylation-related mRNAs in RA for its clinical application. The discovery of m6A methylation-modifying genes in this study may provide a fresh perspective on the development of drugs for RA treatment. High-throughput sequencing combined with methylated RNA immunoprecipitation (MeRIP-seq) and RNA sequencing were used to assess whole-transcriptome m6A modifications in the synovium of patients with RA. The relationship between m6A-modified target genes and RA inflammation and macrophages was determined. The expression of the m6A-modified significant transcript-enriched inflammatory signaling pathway was assessed through animal experiments. Differentially expressed m6A genes were correlated with macrophage activation involved in immune response, vascular endothelium, MAPK signaling pathway, PI3K - Akt signaling pathway, and other inflammatory processes. Furthermore, combined analysis with m6A-seq and RNA-seq revealed 120 genes with significant changes in both m6A modification and mRNA expression. We selected the top 3 candidate mRNAs that were upregulated and downregulated simultaneously. The expression of phosphatase and tensin homolog deleted on chromosome ten (PTEN) mRNA and protein in RA patients was lower than that in healthy control (HC). SHC-binding protein 1 (SHCBP1) and neurexophilin-3 (NXPH3) mRNA expressions were increased in RA patients. The expression of M1 macrophages was increased in RA patients. RA markers are such as rheumatoid factor (RF) and peptide containing citrulline (CCP). Further animal experiments showed that the expression of synovial MAPK, PI3K, and Akt1 proteins in the RA model was increased, and the PTEN, p-PTEN protein expression was decreased. PI3K, Akt1, PTEN, and p-PTEN were correlated to RA joint inflammation. This study revealed a unique pattern of differential m6A methylation modifications in RA and concluded that m6A modification is related to the occurrence of RA synovial inflammation.
Collapse
Affiliation(s)
- Lei Wan
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230038, China
- Key Laboratory of Xin'an Medical Education Ministry, Hefei 230038, China
| | - Jian Liu
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230038, China
- Key Laboratory of Xin'an Medical Education Ministry, Hefei 230038, China
| | - Chuanbing Huang
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230038, China
| | - Ziheng Zhu
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230038, China
| | - Kun Wang
- Key Laboratory of Xin'an Medical Education Ministry, Hefei 230038, China
- College of Traditional Chinese Medicine, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Guanghan Sun
- College of Traditional Chinese Medicine, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Lei Zhu
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230038, China
| | - Zhongxiang Hu
- The First Affiliated Hospital of University of Science and Technology of China, Hefei 230000, China
| |
Collapse
|
9
|
Ansari SA, Dantoft W, Ruiz-Orera J, Syed AP, Blachut S, van Heesch S, Hübner N, Uhlenhaut NH. Integrative analysis of macrophage ribo-Seq and RNA-Seq data define glucocorticoid receptor regulated inflammatory response genes into distinct regulatory classes. Comput Struct Biotechnol J 2022; 20:5622-5638. [PMID: 36284713 PMCID: PMC9582734 DOI: 10.1016/j.csbj.2022.09.042] [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: 07/15/2022] [Revised: 09/28/2022] [Accepted: 09/28/2022] [Indexed: 11/03/2022] Open
Abstract
Glucocorticoids such as dexamethasone (Dex) are widely used to treat both acute and chronic inflammatory conditions. They regulate immune responses by dampening cell-mediated immunity in a glucocorticoid receptor (GR)-dependent manner, by suppressing the expression of pro-inflammatory cytokines and chemokines and by stimulating the expression of anti-inflammatory mediators. Despite its evident clinical benefit, the mechanistic underpinnings of the gene regulatory networks transcriptionally controlled by GR in a context-specific manner remain mysterious. Next generation sequencing methods such mRNA sequencing (RNA-seq) and Ribosome profiling (ribo-seq) provide tools to investigate the transcriptional and post-transcriptional mechanisms that govern gene expression. Here, we integrate matched RNA-seq data with ribo-seq data from human acute monocytic leukemia (THP-1) cells treated with the TLR4 ligand lipopolysaccharide (LPS) and with Dex, to investigate the global transcriptional and translational regulation (translational efficiency, ΔTE) of Dex-responsive genes. We find that the expression of most of the Dex-responsive genes are regulated at both the transcriptional and the post-transcriptional level, with the transcriptional changes intensified on the translational level. Overrepresentation pathway analysis combined with STRING protein network analysis and manual functional exploration, identified these genes to encode immune effectors and immunomodulators that contribute to macrophage-mediated immunity and to the maintenance of macrophage-mediated immune homeostasis. Further research into the translational regulatory network underlying the GR anti-inflammatory response could pave the way for the development of novel immunomodulatory therapeutic regimens with fewer undesirable side effects.
Collapse
Affiliation(s)
- Suhail A. Ansari
- Institute for Diabetes and Endocrinology (IDE), Helmholtz Center Munich (HMGU) and German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Widad Dantoft
- Institute for Diabetes and Endocrinology (IDE), Helmholtz Center Munich (HMGU) and German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Jorge Ruiz-Orera
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Afzal P. Syed
- Institute for Diabetes and Endocrinology (IDE), Helmholtz Center Munich (HMGU) and German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Susanne Blachut
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Sebastiaan van Heesch
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands
| | - Norbert Hübner
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany,Charite-Universitätsmedizin Berlin, Berlin, Germany
| | - Nina Henriette Uhlenhaut
- Institute for Diabetes and Endocrinology (IDE), Helmholtz Center Munich (HMGU) and German Center for Diabetes Research (DZD), Neuherberg, Germany,Metabolic Programming, School of Life Sciences Weihenstephan, ZIEL – Institute for Food and Health, Technical University of Munich (TUM), Freising, Germany,Corresponding author.
| |
Collapse
|
10
|
Wang X, Xu S, Chen D. Reply to the Comment on "The m6A Reader IGF2BP2 Regulates Macrophage Phenotypic Activation and Inflammatory Diseases by Stabilizing TSC1 and PPARγ". ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2201452. [PMID: 35821566 PMCID: PMC9443459 DOI: 10.1002/advs.202201452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 05/21/2022] [Indexed: 06/15/2023]
Affiliation(s)
- Xia Wang
- Laboratory of Translational GastroenterologyDepartment of GastroenterologyQilu HospitalShandong UniversityJinan250012China
| | - Shuai Xu
- Department of Obstetrics and GynecologyThe Second HospitalCheeloo College of MedicineShandong UniversityJinan250033China
| | - Dawei Chen
- Laboratory of Medical ChemistryInterdisciplinary Cluster for Applied GenoproteomicsGIGA‐Stem CellsFaculty of MedicineUniversity of LiègeCHU, Sart‐TilmanLiège4000Belgium
| |
Collapse
|