1
|
Chen J, Sun HW, Wang RZ, Zhang YF, Li WJ, Wang YK, Wang H, Jia MM, Xu QX, Zhuang H, Xue N. Glutamate promotes CCL2 expression to recruit tumor-associated macrophages by restraining EZH2-mediated histone methylation in hepatocellular carcinoma. Oncoimmunology 2025; 14:2497172. [PMID: 40271976 PMCID: PMC12026252 DOI: 10.1080/2162402x.2025.2497172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 04/02/2025] [Accepted: 04/21/2025] [Indexed: 04/25/2025] Open
Abstract
Glutamate is well-known as metabolite for maintaining the energy and redox homeostasis in cancer, moreover it is also the primary excitatory neurotransmitter in the central nervous system. However, whether glutamatergic signaling can regulate hepatocellular carcinoma (HCC) progression and the specific regulatory mechanisms are unknown. In the present study, we found that glutamate and its receptor NMDAR2B were significantly elevated in HCC patients, which predicts poor prognosis. Glutamate could upregulate CCL2 expression on hepatoma cells and further enhance the capability of tumor cells to recruit tumor-associated macrophages (TAMs). Mechanistically, glutamate could facilitate CCL2 expression through NMDAR pathway by decreasing the expression of EZH2, which regulates the H3K27me3 levels on the CCL2 promoter, rather than affecting DNA methylation. Moreover, inhibiting glutamate pathway with MK801 could significantly delay tumor growth, with reduced TAMs in implanted Hepa1-6 mouse HCC models. Our work suggested that glutamate could induce CCL2 expression to promote TAM infiltration by negatively regulating EZH2 levels in hepatoma cells, which might serve as a potential prognostic marker and a therapeutic target for HCC patients.
Collapse
Affiliation(s)
- Jing Chen
- Department of Orthopaedics, Department of Clinical Laboratory, The Affiliated Cancer Hospital of Zhengzhou University&Henan Cancer Hospital, Zhenghou, China
| | - Hong-Wei Sun
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai Clinical Medical College of Jinan University (Zhuhai People’s Hospital), Zhuhai, China
| | - Run-Zheng Wang
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yun-Fei Zhang
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wen-Jiao Li
- Department of Orthopaedics, Department of Clinical Laboratory, The Affiliated Cancer Hospital of Zhengzhou University&Henan Cancer Hospital, Zhenghou, China
| | - Yong-Kui Wang
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hao Wang
- Department of Orthopaedics, Department of Clinical Laboratory, The Affiliated Cancer Hospital of Zhengzhou University&Henan Cancer Hospital, Zhenghou, China
| | - Miao-Miao Jia
- Department of Orthopaedics, Department of Clinical Laboratory, The Affiliated Cancer Hospital of Zhengzhou University&Henan Cancer Hospital, Zhenghou, China
| | - Qing-Xia Xu
- Department of Orthopaedics, Department of Clinical Laboratory, The Affiliated Cancer Hospital of Zhengzhou University&Henan Cancer Hospital, Zhenghou, China
| | - Hao Zhuang
- Department of Orthopaedics, Department of Clinical Laboratory, The Affiliated Cancer Hospital of Zhengzhou University&Henan Cancer Hospital, Zhenghou, China
| | - Ning Xue
- Department of Orthopaedics, Department of Clinical Laboratory, The Affiliated Cancer Hospital of Zhengzhou University&Henan Cancer Hospital, Zhenghou, China
| |
Collapse
|
2
|
Dai Q, Ma PF, Tian J, Zhang Z, Fang Q, Lin ZY, Wu LL, Li XM. Methyltransferase-like factor 14 pathway and its relationship with pathological stages in patients undergoing surgery for colorectal cancer. World J Gastrointest Surg 2025; 17:101674. [DOI: 10.4240/wjgs.v17.i5.101674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 02/03/2025] [Accepted: 03/18/2025] [Indexed: 05/23/2025] Open
Abstract
BACKGROUND Epigenetic involvement of methyltransferase-like factor 14 (METTL14) in tumor development has not been clearly defined.
AIM To investigate METTL14 expression and its relationship with pathological stage in patients undergoing radical surgery for colorectal cancer (CRC).
METHODS This retrospective study included 80 patients with CRC who were admitted to the Third Hospital of Hefei and the Western District of the First Affiliated Hospital of Anhui Medical University between June 2021 and June 2024. These patients were selected for treatment. Lesions and adjacent tissues were collected from these patients, and METTL14 expression was assessed using immunohistochemistry. Expression levels of METTL14 were compared across different tissue samples. Additionally, we evaluated METTL14 expression in patients with varying pathological characteristics using statistical methods such as χ2 tests and analysis of variance to identify significant differences.
RESULTS The positivity rate of METTL14 in tumor tissues was significantly lower than that in adjacent tissues (30% vs 60%, P < 0.05). Conversely, the negative expression rate of METTL14 was higher in tumor tissues compared to adjacent tissues (P < 0.05). The positive expression of METTL14 mRNA did not differ by age, sex, tumor tissue classification, tumor diameter, or tumor location (P > 0.05). However, the positive expression rate of METTL14 was significantly lower in patients with lymph node metastasis, invasion depth T3 + T4, and tumor, node, and metastasis (TNM) stage III/IV compared to those without lymph node metastasis, invasion depth T1 + T2, and TNM stage I (P < 0.05). Specifically, METTL14 mRNA expression was significantly lower in patients with lymph node metastasis (0.51 ± 0.12 vs 1.23 ± 0.25, P < 0.001), invasion depth T3 + T4 (0.48 ± 0.15 vs 1.18 ± 0.21, P < 0.001), and TNM stage III/IV (0.45 ± 0.13 vs 1.20 ± 0.22, P < 0.001) compared to those with no lymph node metastasis, invasion depth T1 + T2, and TNM stage I, respectively.
CONCLUSION In CRC, low positive METTL14 expression is closely correlated with lymph node metastasis, invasion depth T3 + T4, and TNM stage, indicating the malignant biological behavior of rectal cancer.
Collapse
Affiliation(s)
- Qun Dai
- Department of Pathology, The Third People's Hospital of Hefei, Hefei Third Clinical College of Anhui Medical University, Hefei 230022, Anhui Province, China
| | - Peng-Fei Ma
- Department of Gastroenterology, Children’s Hospital of Fudan University at Anhui, Anhui Provincial Children’s Hospital, Hefei 230022, Anhui Province, China
| | - Jing Tian
- Department of Pathology, The Third People's Hospital of Hefei, Hefei Third Clinical College of Anhui Medical University, Hefei 230022, Anhui Province, China
| | - Zhan Zhang
- Department of Pathology, South District, The First Affiliated Hospital of Anhui Medical University, Feixi 231200, Anhui Province, China
| | - Qin Fang
- Department of Pathology, The Third People's Hospital of Hefei, Hefei Third Clinical College of Anhui Medical University, Hefei 230022, Anhui Province, China
| | - Zi-Yu Lin
- Department of Pathology, The Third People's Hospital of Hefei, Hefei Third Clinical College of Anhui Medical University, Hefei 230022, Anhui Province, China
| | - Li-Li Wu
- Department of Pathology, The Third People's Hospital of Hefei, Hefei Third Clinical College of Anhui Medical University, Hefei 230022, Anhui Province, China
| | - Xue-Min Li
- Department of Pathology, The Third People's Hospital of Hefei, Hefei Third Clinical College of Anhui Medical University, Hefei 230022, Anhui Province, China
| |
Collapse
|
3
|
Yao Z, Sun L, Gao Y, Su Y, He B, Ge Y, Yang C, Jia X, Jiao A, Sun C, Zhang B. The m 6A demethylase FTO controls Th1 differentiation and immunity against infections. Mol Immunol 2025; 183:172-181. [PMID: 40378511 DOI: 10.1016/j.molimm.2025.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2025] [Revised: 05/05/2025] [Accepted: 05/05/2025] [Indexed: 05/19/2025]
Abstract
Antigen-specific effector CD4+ T cells are critical for defense against exogenous pathogens. However, the epigenetic mechanisms underlying CD4+ T cell immune responses, particularly RNA modifications, remain incompletely understood. In this study, we employed a T cell-specific deletion of the fat mass and obesity-associated protein (FTO), a key N6-methyladenosine (m6A) demethylase, to elucidate its role in CD4+ T cell mediated immunity. Our findings demonstrate that FTO is essential for maintaining CD4+ T cell immune responses and protective functions. Specifically, FTO deficiency restricts the expansion of CD4+ T helper (Th)1 effector cells following antigen challenge and results in decreased expression of T-bet and IFN-γ in Th1 cells. Additionally, FTO deficient CD4+ T cells exhibit impaired pathogen elimination. Collectively, our study reveals a novel epigenetic regulatory mechanism in supporting CD4+ T cell differentiation, providing new insights into the post-transcriptional regulation of CD4+ T cell immunity and highlighting the potential for therapeutic strategies.
Collapse
Affiliation(s)
- Zhihong Yao
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Faculty of Clinical Medicine, Hanzhong Vocational and Technical College, Hanzhong 723002, China
| | - Lina Sun
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi 710061, China; Key Laboratory for ImmunoHealth of Shaanxi Province, Xi'an, Shannxi 710061, China
| | - Yang Gao
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Department of Kidney Transplantation, Hospital of Nephropathy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China; Institute of Organ Transplantation, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Yanhong Su
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi 710061, China; Key Laboratory for ImmunoHealth of Shaanxi Province, Xi'an, Shannxi 710061, China
| | - Boxiao He
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi 710061, China; Key Laboratory for ImmunoHealth of Shaanxi Province, Xi'an, Shannxi 710061, China
| | - Yao Ge
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Chen Yang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Xiaoxuan Jia
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Anjun Jiao
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi 710061, China; Key Laboratory for ImmunoHealth of Shaanxi Province, Xi'an, Shannxi 710061, China
| | - Chenming Sun
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi 710061, China; Key Laboratory for ImmunoHealth of Shaanxi Province, Xi'an, Shannxi 710061, China
| | - Baojun Zhang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi 710061, China; Key Laboratory for ImmunoHealth of Shaanxi Province, Xi'an, Shannxi 710061, China.
| |
Collapse
|
4
|
Teng M, Gu Y, Wang T, Wang Y, Ma Z, Li Y, Fan Y, Wan Q, Li Y. Transforming the Tumor Microenvironment: An Outstanding AIE-Active Photosensitizer to Boost the Effectiveness of Immunotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025:e2503355. [PMID: 40351086 DOI: 10.1002/smll.202503355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2025] [Revised: 04/26/2025] [Indexed: 05/14/2025]
Abstract
Immunotherapy, currently the most promising therapeutic approach for cancer, has shown significant efficacy. However, its clinical effectiveness is often constrained by such factors as tumor heterogeneity, the abundance of M2 macrophages, tumor-vascular hypoxia, and the immunosuppressive microenvironment created by immune checkpoint (IC) complexes. In this work, an effective photosensitizer (TSPA) with aggregation-induced emission (AIE) nature is adopted to counter above limitations. The synthesized TSPA demonstrated potent efficacy in eradicating primary tumors because of their effective generation reactive oxygen species (ROS) after undergoing photodynamic therapy (PDT) process. Moreover, TSPA can improve hypoxic conditions in tumor by normalizing blood vessels, and can instigate immunogenic cell death (ICD), thus stimulating immune cell activation. TSPA demonstrates the ability to reprogram M2 tumor-associated macrophages (TAMs) into the anti-tumor M1 phenotype, thereby increasing the infiltration of M1 macrophages within the tumor. This procedure notably ameliorates the immune microenvironment, effectively suppressing the long-term metastasis of breast cancer (BC). This research notably enhances the efficiency of tumor immunotherapy and is anticipated to emerge as a new strategy for improving the tumor's immunosuppressive microenvironment and overcoming immune evasion.
Collapse
Affiliation(s)
- Muzhou Teng
- Gansu Provincial Maternity and Child-Care Hospital(Gansu Provincial Central Hospital), Lanzhou, 730050, China
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730000, China
| | - Yanmei Gu
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730000, China
| | - Tongxin Wang
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730000, China
| | - Yingying Wang
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730000, China
| | - Zihang Ma
- School of Materials Science and Engineering, Nanchang Hangkong University, Nanchang, 330063, China
| | - Yirong Li
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730000, China
| | - Yitao Fan
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730000, China
| | - Qing Wan
- School of Materials Science and Engineering, Nanchang Hangkong University, Nanchang, 330063, China
- AIE institute, South China University of Technology, Guangzhou, 510640, China
| | - Yumin Li
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730000, China
| |
Collapse
|
5
|
Viiklepp K, Knuutila JS, Nissinen L, Siljamäki E, Rappu P, Suwal U, Pellinen T, Kallajoki M, Meri S, Heino J, Kähäri VM, Riihilä P. Expression of C1q by Macrophages and Fibroblasts in Tumor Microenvironment Is Associated with Progression and Metastasis of Cutaneous Squamous Cell Carcinoma. J Invest Dermatol 2025:S0022-202X(25)00446-4. [PMID: 40311866 DOI: 10.1016/j.jid.2025.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 03/10/2025] [Accepted: 04/02/2025] [Indexed: 05/03/2025]
Abstract
Cutaneous squamous cell carcinoma (cSCC) is the most common metastatic skin cancer, with poor prognosis for metastatic cases. We demonstrated previously that cSCC cells in culture express C1r and C1s components of the complement C1qr2s2 complex but not C1q. In this study, significantly higher mRNA levels of C1QA, C1QB, and C1QC variants 1 and 2 were found in cSCC tumors than in normal skin. Analysis of single-cell RNA-sequencing data of cSCC revealed expression of mRNAs for C1QA, C1QB, and C1QC in macrophages and activated fibroblasts. C1q staining was detected on the surface of cSCC tumor cells, in peritumoral and intratumoral macrophages, and in peritumoral activated fibroblasts using immunohistochemistry and multiplexed immunofluorescence. Expression was higher in cSCCs than in normal skin, actinic keratoses, and cSCC in situ. C1q production was induced in 3-dimensional spheroid cocultures of cSCC cells, fibroblasts, and macrophages. C1q stimulated the growth of cSCC cells in culture. C1q expression was significantly more prevalent in metastatic primary cSCCs and in metastases than in non-metastatic cSCCs. High C1q expression in cSCC correlated with poor prognosis. These findings provide evidence for macrophage- and fibroblast-derived C1q in the progression of cSCC. They also suggest stromal C1q as a marker for cSCC metastasis and poor prognosis.
Collapse
Affiliation(s)
- Kristina Viiklepp
- Department of Dermatology, University of Turku and Turku University Hospital, Turku, Finland; FICAN West Cancer Centre Laboratory, University of Turku and Turku University Hospital, Turku, Finland
| | - Jaakko S Knuutila
- Department of Dermatology, University of Turku and Turku University Hospital, Turku, Finland; FICAN West Cancer Centre Laboratory, University of Turku and Turku University Hospital, Turku, Finland
| | - Liisa Nissinen
- Department of Dermatology, University of Turku and Turku University Hospital, Turku, Finland; FICAN West Cancer Centre Laboratory, University of Turku and Turku University Hospital, Turku, Finland
| | - Elina Siljamäki
- MediCity Research Laboratory, University of Turku, Turku, Finland; Department of Life Technologies, University of Turku, Turku, Finland; InFLAMES Research Flagship, University of Turku, Turku, Finland
| | - Pekka Rappu
- MediCity Research Laboratory, University of Turku, Turku, Finland; Department of Life Technologies, University of Turku, Turku, Finland; InFLAMES Research Flagship, University of Turku, Turku, Finland
| | - Ujjwal Suwal
- MediCity Research Laboratory, University of Turku, Turku, Finland; Department of Life Technologies, University of Turku, Turku, Finland; InFLAMES Research Flagship, University of Turku, Turku, Finland
| | - Teijo Pellinen
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Markku Kallajoki
- Department of Pathology, University of Turku and Turku University Hospital, Turku, Finland
| | - Seppo Meri
- Department of Bacteriology and Immunology, University of Helsinki, Helsinki, Finland; Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Jyrki Heino
- MediCity Research Laboratory, University of Turku, Turku, Finland; Department of Life Technologies, University of Turku, Turku, Finland; InFLAMES Research Flagship, University of Turku, Turku, Finland
| | - Veli-Matti Kähäri
- Department of Dermatology, University of Turku and Turku University Hospital, Turku, Finland; FICAN West Cancer Centre Laboratory, University of Turku and Turku University Hospital, Turku, Finland.
| | - Pilvi Riihilä
- Department of Dermatology, University of Turku and Turku University Hospital, Turku, Finland; FICAN West Cancer Centre Laboratory, University of Turku and Turku University Hospital, Turku, Finland.
| |
Collapse
|
6
|
Liu L, Ge D, Lin Y, Han Z, Zhao H, Cao L, Wu X, Ma G. Epigenetic regulation in oogenesis and fetal development: insights into m6A modifications. Front Immunol 2025; 16:1516473. [PMID: 40356909 PMCID: PMC12066277 DOI: 10.3389/fimmu.2025.1516473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 04/07/2025] [Indexed: 05/15/2025] Open
Abstract
The unique physiological structure of women has led to a variety of diseases that have attracted the attention of many people in recent years. Disturbances in the reproductive system microenvironment lead to the progression of various female tumours and pregnancy disorders. Numerous studies have shown that epigenetic modifications crucially influence both oogenesis and foetal development. m6A, a modification at the mRNA level, consists of three parts, namely, writers, erasers, and readers, which are involved in several biological functions, such as the nucleation and stabilisation of mRNAs, thereby regulating the development of reproductive system diseases. In this manuscript, we delineate the constituents of m6A, their biological roles, and advancements in understanding m6A within the maternal-foetal immunological context. In addition, we summarise the mechanism of m6A in gynaecological diseases and provide a new perspective for targeting m6A to delay the progression of reproductive system diseases in clinical practice.
Collapse
Affiliation(s)
- Lusheng Liu
- Department of Acupuncture and Moxibustion, Shanghai Traditional Chinese Medicine (TCM)-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Clinical Medical College of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Danxia Ge
- Department of Critical Care Medicine, Traditional Chinese Medicine Hospital of, Ningbo, Zhejiang, China
| | - Yumeng Lin
- Nanjing Tongren Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Zhongyu Han
- Department of Acupuncture and Moxibustion, Shanghai Traditional Chinese Medicine (TCM)-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Heng Zhao
- Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Liqin Cao
- Department of Gynecology, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xi Wu
- Department of Gynecology, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guizhi Ma
- Department of Acupuncture and Moxibustion, Shanghai Traditional Chinese Medicine (TCM)-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
7
|
Sun L, Zhang T, Ge Y, Yao Z, Su Y, Wang Q, Chen Y, He B, Ding R, Zhang C, Lan L, Liu R, Ping H, Zhang D, Shi L, Zhou X, Jia X, Sun C, Liang L, Zhang L, Zhang B. FTO controls CD8 + T cell survival and effector response by modulating m 6A methylation of Fas. Cell Death Dis 2025; 16:301. [PMID: 40234389 PMCID: PMC12000336 DOI: 10.1038/s41419-025-07606-z] [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: 12/20/2024] [Revised: 03/19/2025] [Accepted: 03/27/2025] [Indexed: 04/17/2025]
Abstract
Functional CD8+ T cell immunity is essential for immune surveillance and host defense against infection and tumors. Epigenetic mechanisms, particularly RNA modification, in controlling CD8+ T cell immune response is not fully elucidated. Here, by T cell-specific deletion of fat mass and obesity-associated protein (FTO), a critical N6-methyladenosine (m6A) demethylase, we revealed that FTO was indispensable for adequate CD8+ T cell immune response and protective function. FTO ablation led to considerable cell death in activated CD8+ T cells, which was attributed to cell apoptosis. MeRIP-seq analysis revealed an increase in m6A methylation on Fas mRNA in FTO-deficient CD8+ T cells. The loss of FTO promoted Fas expression via enhancing the Fas mRNA stability, which depended on the m6A reader insulin-like growth factor-2 mRNA-biding proteins 3 (IGF2BP3). Mutation of the Fas m6A sites or knockdown IGF2BP3 could normalize the upregulated Fas expression and apoptosis levels caused by FTO ablation in CD8+ T cells. Our findings delineate a novel epigenetic regulatory mechanism of FTO-mediated m6A modification in supporting CD8+ T cell survival and effector responses, providing new insights into understanding the post-transcriptional regulation in CD8+ T cell immunological functions and the potential therapeutic intervention.
Collapse
Affiliation(s)
- Lina Sun
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, China
| | - Tianzhe Zhang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, China
| | - Yao Ge
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Zhihong Yao
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Faculty of Clinical Medicine, Hanzhong Vocational and Technical College, Hanzhong, China
| | - Yanhong Su
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, China
| | - Qianhao Wang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, China
| | - Yang Chen
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Boxiao He
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Renyi Ding
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Cangang Zhang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Linbo Lan
- Clinical Teaching and Research Center, School of Nursing, Weinan Vocational and Technical College, Weinan, Shaanxi, China
| | - Ruonan Liu
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Huanxin Ping
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Dan Zhang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Lin Shi
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, China
| | - Xiaobo Zhou
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, China
| | - Xiaoxuan Jia
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Chenming Sun
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, China.
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China.
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China.
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, China.
| | - Lingli Liang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, PR China; Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, PR China.
| | - Lianjun Zhang
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, Jiangsu, China.
- Key Laboratory of Synthetic Biology Regulatory Elements, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, Jiangsu, China.
| | - Baojun Zhang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, China.
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China.
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China.
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, China.
| |
Collapse
|
8
|
Alcindor T, Tankel J, Fiset PO, Pal S, Opu T, Strasser M, Dehghani M, Bertos N, Zuo D, Mueller C, Cools-Lartigue J, Hickeson M, Marcus V, Camilleri-Broet S, Spatz A, Evaristo G, Farag M, Artho G, Elkrief A, Saleh R, Bailey S, Park M, Huang S, Sangwan V, Ferri L. Phase 2 trial of perioperative chemo-immunotherapy for gastro-esophageal adenocarcinoma: The role of M2 macrophage landscape in predicting response. Cell Rep Med 2025; 6:102045. [PMID: 40239627 PMCID: PMC12047487 DOI: 10.1016/j.xcrm.2025.102045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 11/28/2024] [Accepted: 03/06/2025] [Indexed: 04/18/2025]
Abstract
We present the clinical results of a phase 2 trial combining neoadjuvant docetaxel, cisplatin, 5 Flourouracil, and the PD-L1 inhibitor avelumab in locally advanced gastro-esophageal adenocarcinoma (GEA). Fifty-one patients receive neoadjuvant therapy with 50 proceeding to surgery. Grade 3-4 adverse events occur in 40%; complete/major pathological response is found in 7/50 (14%) and 9/50 (18%), with 2-year disease-free survival of 67.5%. There is no correlation between tumor regression and PD-L1 or mismatch repair (MMR) status. Multiplex immunohistochemistry and longitudinal single-cell transcriptomic profiling reveal alterations in certain innate immune cell populations, particularly noting an M2-tumor-associated macrophage (M2-TAM) proliferation in non-responding tumors. These findings describe the effective nature of this treatment regimen for GEA and reveal associated features of the inflammatory milieux associated with response to chemo-immunotherapy. The specific character of the inflammatory environment in non-responders may, in the future, help personalize treatment. This study was registered at ClinicalTrials.gov (NCT03288350).
Collapse
Affiliation(s)
- Thierry Alcindor
- Department of Medicine, McGill University Health Centre, Montreal, QC, Canada; Center for Innovative Medicine, McGill University Health Centre, Montreal, QC, Canada.
| | - James Tankel
- Department of Surgery, McGill University Health Centre, Montreal, QC, Canada
| | - Pierre-Olivier Fiset
- Division of Pathology, Department of Clinical Laboratory Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Sanjima Pal
- Department of Surgery, McGill University Health Centre, Montreal, QC, Canada
| | - Touhid Opu
- Center for Innovative Medicine, McGill University Health Centre, Montreal, QC, Canada
| | | | - Mehrnoush Dehghani
- Department of Surgery, McGill University Health Centre, Montreal, QC, Canada
| | - Nicholas Bertos
- Department of Surgery, McGill University Health Centre, Montreal, QC, Canada
| | - Dongmei Zuo
- Center for Innovative Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Carmen Mueller
- Department of Surgery, McGill University Health Centre, Montreal, QC, Canada
| | | | - Marc Hickeson
- Department of Nuclear Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Victoria Marcus
- Department of Surgery, McGill University Health Centre, Montreal, QC, Canada
| | - Sophie Camilleri-Broet
- Division of Pathology, Department of Clinical Laboratory Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Alan Spatz
- Division of Pathology, Department of Clinical Laboratory Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Gertruda Evaristo
- Division of Pathology, Department of Clinical Laboratory Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Mina Farag
- Division of Pathology, Department of Clinical Laboratory Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Giovanni Artho
- Department of Diagnostic Radiology, McGill University Health Centre, Montreal, QC, Canada
| | - Arielle Elkrief
- Department of Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Ramy Saleh
- Department of Medicine, McGill University Health Centre, Montreal, QC, Canada; Center for Innovative Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Swneke Bailey
- Department of Surgery, McGill University Health Centre, Montreal, QC, Canada
| | - Morag Park
- Center for Innovative Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Sui Huang
- Institute for Systems Biology, Seattle, WA, USA
| | - Veena Sangwan
- Department of Surgery, McGill University Health Centre, Montreal, QC, Canada
| | - Lorenzo Ferri
- Department of Surgery, McGill University Health Centre, Montreal, QC, Canada.
| |
Collapse
|
9
|
Fan B, Chen G, Huang S, Li Y, Nabil ZUH, Yang Z. Summary of the mechanism of ferroptosis regulated by m6A modification in cancer progression. Front Cell Dev Biol 2025; 13:1507171. [PMID: 40271153 PMCID: PMC12014555 DOI: 10.3389/fcell.2025.1507171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 03/25/2025] [Indexed: 04/25/2025] Open
Abstract
The most common form of internal RNA modification in eukaryotes is called n6-methyladenosine (m6A) methylation. It has become more and more well-known as a research issue in recent years since it alters RNA metabolism and is involved in numerous biological processes. Currently, m6A alteration offers new opportunities in clinical applications and is intimately linked to carcinogenesis. Ferroptosis-a form of iron-dependent, lipid peroxidation-induced regulated cell death-was discovered. In the development of cancer, it has become an important factor. According to newly available data, ferroptosis regulates tumor growth, and cancer exhibits aberrant m6A levels in crucial ferroptosis regulatory components. On the other hand, m6A has multiple roles in the development of tumors, and the relationship between m6A-modified ferroptosis and malignancies is quite intricate. In this review, we first give a thorough review of the regulatory and functional roles of m6A methylation, focusing on the molecular processes of m6A through the regulation of ferroptosis in human cancer progression and metastasis, which are strongly associated to cancer initiation, progression, and drug resistance. Therefore, it is crucial to clarify the relationship between m6A-mediated regulation of ferroptosis in cancer progression, providing a new strategy for cancer treatment with substantial clinical implications.
Collapse
Affiliation(s)
| | | | | | | | | | - Zuozhang Yang
- Bone and Soft Tissue Tumors Research Centre of Yunnan Province, Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital), Kunming, Yunnan, China
| |
Collapse
|
10
|
Ding YP, Liu CC, Yu KD. RNA modifications in the tumor microenvironment: insights into the cancer-immunity cycle and beyond. Exp Hematol Oncol 2025; 14:48. [PMID: 40176140 PMCID: PMC11963313 DOI: 10.1186/s40164-025-00648-1] [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: 11/19/2024] [Accepted: 03/24/2025] [Indexed: 04/04/2025] Open
Abstract
The chemical modification of biological molecules is a critical regulatory mechanism for controlling molecular functions. Although research has long focused on DNA and proteins, RNA modifications have recently attracted substantial interest with the advancement in detection technologies. In oncology, many studies have identified dysregulated RNA modifications including m6A, m1A, m5C, m7G, pseudouridylation and A to I editing, leading to disrupted downstream pathways. As the concept of the tumor microenvironment has gained prominence, studies have increasingly examined the role of RNA modifications in this context, focusing on interactions among cancer cells, immune cells, stromal cells, and other components. Here we review the RNA modifications in the tumor microenvironment through the perspective of the Cancer-Immunity Cycle. The extracellular RNA modifications including exosomes and influence of microbiome in RNA modifications are potential research questions. Additionally, RNA modifying enzymes including FTO, ALKBH5, METTL3, PUS7 are under investigation as potential biomarkers and targets for combination with immunotherapies. ADCs and mimetics of modified RNA could be potential novel drugs. This review discusses the regulatory roles of RNA modifications within the tumor microenvironment.
Collapse
Affiliation(s)
- You-Peng Ding
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Department of Oncology, Shanghai Medical College, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
| | - Cui-Cui Liu
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Department of Oncology, Shanghai Medical College, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
| | - Ke-Da Yu
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Department of Oncology, Shanghai Medical College, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
11
|
Zhao L, Guo J, Xu S, Duan M, Liu B, Zhao H, Wang Y, Liu H, Yang Z, Yuan H, Jiang X, Jiang X. Abnormal changes in metabolites caused by m 6A methylation modification: The leading factors that induce the formation of immunosuppressive tumor microenvironment and their promising potential for clinical application. J Adv Res 2025; 70:159-186. [PMID: 38677545 PMCID: PMC11976433 DOI: 10.1016/j.jare.2024.04.016] [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: 02/18/2024] [Revised: 04/14/2024] [Accepted: 04/14/2024] [Indexed: 04/29/2024] Open
Abstract
BACKGROUND N6-methyladenosine (m6A) RNA methylation modifications have been widely implicated in the metabolic reprogramming of various cell types within the tumor microenvironment (TME) and are essential for meeting the demands of cellular growth and maintaining tissue homeostasis, enabling cells to adapt to the specific conditions of the TME. An increasing number of research studies have focused on the role of m6A modifications in glucose, amino acid and lipid metabolism, revealing their capacity to induce aberrant changes in metabolite levels. These changes may in turn trigger oncogenic signaling pathways, leading to substantial alterations within the TME. Notably, certain metabolites, including lactate, succinate, fumarate, 2-hydroxyglutarate (2-HG), glutamate, glutamine, methionine, S-adenosylmethionine, fatty acids and cholesterol, exhibit pronounced deviations from normal levels. These deviations not only foster tumorigenesis, proliferation and angiogenesis but also give rise to an immunosuppressive TME, thereby facilitating immune evasion by the tumor. AIM OF REVIEW The primary objective of this review is to comprehensively discuss the regulatory role of m6A modifications in the aforementioned metabolites and their potential impact on the development of an immunosuppressive TME through metabolic alterations. KEY SCIENTIFIC CONCEPTS OF REVIEW This review aims to elaborate on the intricate networks governed by the m6A-metabolite-TME axis and underscores its pivotal role in tumor progression. Furthermore, we delve into the potential implications of the m6A-metabolite-TME axis for the development of novel and targeted therapeutic strategies in cancer research.
Collapse
Affiliation(s)
- Liang Zhao
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China; Department of Colorectal Anal Surgery, Shenyang Coloproctology Hospital, Shenyang 110002, China.
| | - Junchen Guo
- Department of Radiology, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| | - Shasha Xu
- Department of Gastroendoscopy, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| | - Meiqi Duan
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| | - Baiming Liu
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| | - He Zhao
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| | - Yihan Wang
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| | - Haiyang Liu
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| | - Zhi Yang
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| | - Hexue Yuan
- Department of Colorectal Anal Surgery, Shenyang Coloproctology Hospital, Shenyang 110002, China.
| | - Xiaodi Jiang
- Department of Infectious Disease, Shengjing Hospital of China Medical University, Shenyang 110020, China.
| | - Xiaofeng Jiang
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| |
Collapse
|
12
|
Xiong Q, Zhang Y, Zheng Y, Zhu Q. Regulation and application of m 6A modification in tumor immunity. SCIENCE CHINA. LIFE SCIENCES 2025; 68:974-993. [PMID: 39648245 DOI: 10.1007/s11427-024-2648-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 06/11/2024] [Indexed: 12/10/2024]
Abstract
The m6A modification is an RNA modification that impacts various processes of RNA molecules, including transcription, splicing, stability, and translation. Recently, researchers have discovered that the presence of m6A modification can influence the interaction between tumor cells and immune cells and also play a role in regulating the expression of immune response-related genes. Additionally, m6A modification is intricately involved in the regulation of tumor immune evasion and drug resistance. Specifically, certain tumor cells can manipulate the gene expression through m6A modification to evade immune system attacks. Therefore, it might be possible to enhance tumor immune surveillance and improve the effectiveness of immune-based therapies by manipulating m6A modification. This review systematically discusses the role of m6A modification in tumor immunity, specifically highlighting its regulation of immune cells and immune-related genes in tumor cells. Furthermore, we explore the potential of m6A modification inhibitors as anti-cancer therapies and the significance of m6A regulatory factors in predicting the efficacy of tumor immune therapy.
Collapse
Affiliation(s)
- Qunli Xiong
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yaguang Zhang
- Laboratory of Gastrointestinal Tumor Epigenetics and Genomics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ying Zheng
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qing Zhu
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
13
|
Xu L, Shen T, Li Y, Wu X. The Role of M 6A Modification in Autoimmunity: Emerging Mechanisms and Therapeutic Implications. Clin Rev Allergy Immunol 2025; 68:29. [PMID: 40085180 DOI: 10.1007/s12016-025-09041-6] [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] [Accepted: 03/03/2025] [Indexed: 03/16/2025]
Abstract
N6-methyladenosine (m6A), a prevalent and essential RNA modification, serves a key function in driving autoimmune disease pathogenesis. By modulating immune cell development, activation, migration, and polarization, as well as inflammatory pathways, m6A is crucial in forming innate defenses and adaptive immunity. This article provides a comprehensive overview of m6A modification features and reveals how its dysregulation affects the intensity and persistence of immune responses, disrupts immune tolerance, exacerbates tissue damage, and promotes the development of autoimmunity. Specific examples include its contributions to systemic autoimmune disorders like lupus and rheumatoid arthritis, as well as conditions that targeting specific organs like multiple sclerosis and type 1 diabetes. Furthermore, this review explores the therapeutic promise of target m6A-related enzymes ("writers," "erasers," and "readers") and summarizes recent advances in intervention strategies. By focusing on the mechanistic and therapeutic implications of m6A modification, this review sheds light on its role as a promising tool for both diagnosis and treatment in autoimmune disorders, laying the foundation for advancements in customized medicine.
Collapse
Affiliation(s)
- Liyun Xu
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Tian Shen
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Yongzhen Li
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.
| | - Xiaochuan Wu
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.
| |
Collapse
|
14
|
Ji W, Fang Y, Chen L, Zheng Y, Pei Y, Mei C, Zhou M. Pan-cancer characterization of m6A-mediated regulation of T cell exhaustion dynamics and clinical relevancies in human cancers. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102465. [PMID: 39995977 PMCID: PMC11847731 DOI: 10.1016/j.omtn.2025.102465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 01/22/2025] [Indexed: 02/26/2025]
Abstract
T cell exhaustion (TEX) is a major barrier to effective immunotherapy. The role of N6-methyladenosine (m6A) modification in regulating immune cell function has been recognized, but its impact on TEX dynamics across cancer types and clinical outcomes remains unclear. Here, we conducted a pan-cancer analysis integrating multi-omics data from cell lines, single-cell RNA sequencing, and pan-cancer and immunotherapy datasets to explore the dynamic interplay between m6A modification and TEX. We found that m6A modification influences key TEX-associated genes at both the cellular and single-cell levels, with distinct expression patterns across the exhaustion spectrum. Based on m6A-TEX interactions, three pan-cancer subtypes were identified, each with unique molecular profiles, immune phenotypes, and survival outcomes. The TexLm6AL subtype, characterized by low m6A activity and low TEX, correlated with high immune infiltration, increased cytolytic activity, and favorable survival, whereas the TexLm6AH and TexHm6AH subtypes with higher m6A activity were associated with poorer survival. Multivariate analysis confirmed the prognostic value of this classification independent of traditional clinical factors. Moreover, m6A-TEX crosstalk influenced responses to immune checkpoint blockade therapies. Our findings provide novel insights into the role of m6A in TEX regulation and underscore the potential of m6A regulators as biomarkers and therapeutic targets for advancing cancer immunotherapy.
Collapse
Affiliation(s)
- Weiping Ji
- Department of Genaral Surgery, School of Biomedical Engineering, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital, Wenzhou Medical University, Zhejiang, P.R. China
| | - Ye Fang
- Department of Genaral Surgery, School of Biomedical Engineering, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital, Wenzhou Medical University, Zhejiang, P.R. China
| | - Liwei Chen
- Department of Genaral Surgery, School of Biomedical Engineering, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital, Wenzhou Medical University, Zhejiang, P.R. China
| | - Yitong Zheng
- Department of Genaral Surgery, School of Biomedical Engineering, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital, Wenzhou Medical University, Zhejiang, P.R. China
| | - Yifei Pei
- Department of Genaral Surgery, School of Biomedical Engineering, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital, Wenzhou Medical University, Zhejiang, P.R. China
| | - Changqiu Mei
- Department of Genaral Surgery, School of Biomedical Engineering, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital, Wenzhou Medical University, Zhejiang, P.R. China
| | - Meng Zhou
- Department of Genaral Surgery, School of Biomedical Engineering, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital, Wenzhou Medical University, Zhejiang, P.R. China
| |
Collapse
|
15
|
Zhu Q, Zhang R, Zhao Z, Xie T, Sui X. Harnessing phytochemicals: Innovative strategies to enhance cancer immunotherapy. Drug Resist Updat 2025; 79:101206. [PMID: 39933438 DOI: 10.1016/j.drup.2025.101206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 01/18/2025] [Accepted: 01/23/2025] [Indexed: 02/13/2025]
Abstract
Cancer immunotherapy has revolutionized cancer treatment, but therapeutic ineffectiveness-driven by the tumor microenvironment and immune evasion mechanisms-continues to limit its clinical efficacy. This challenge underscores the need to explore innovative approaches, such as multimodal immunotherapy. Phytochemicals, bioactive compounds derived from plants, have emerged as promising candidates for overcoming these barriers due to their immunomodulatory and antitumor properties. This review explores the synergistic potential of phytochemicals in enhancing immunotherapy by modulating immune responses, reprogramming the tumor microenvironment, and reducing immunosuppressive factors. Integrating phytochemicals with conventional immunotherapy strategies represents a novel approach to mitigating resistance and enhancing therapeutic outcomes. For instance, nab-paclitaxel has shown the potential in overcoming resistance to immune checkpoint inhibitors, while QS-21 synergistically enhances the efficacy of tumor vaccines. Furthermore, we highlight recent advancements in leveraging nanotechnology to engineer phytochemicals for improved bioavailability and targeted delivery. These innovations hold great promise for optimizing the clinical application of phytochemicals. However, further large-scale clinical studies are crucial to fully integrate these compounds into immunotherapeutic regimens effectively.
Collapse
Affiliation(s)
- Qianru Zhu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macao
| | - Ruonan Zhang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Ziming Zhao
- State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macao
| | - Tian Xie
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macao; Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, Zhejiang 310015, China.
| | - Xinbing Sui
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macao; Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, Zhejiang 310015, China.
| |
Collapse
|
16
|
Chen X, Yuan Y, Zhou F, Li L, Pu J, Jiang X. m6A RNA methylation: a pivotal regulator of tumor immunity and a promising target for cancer immunotherapy. J Transl Med 2025; 23:245. [PMID: 40022120 PMCID: PMC11871626 DOI: 10.1186/s12967-025-06221-y] [Citation(s) in RCA: 2] [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: 09/27/2024] [Accepted: 02/11/2025] [Indexed: 03/03/2025] Open
Abstract
M6A modification is one of the most common regulatory mechanisms of gene expression in eukaryotic cells, influencing processes such as RNA splicing, degradation, stability, and protein translation. Studies have shown that m6A methylation is closely associated with tumorigenesis and progression, and it plays a key regulatory role in tumor immune responses. m6A modification participates in regulating the differentiation and maturation of immune cells, as well as related anti-tumor immune responses. In the tumor microenvironment, m6A modification can also affect immune cell recruitment, activation, and polarization, thereby promoting or inhibiting tumor cell proliferation and metastasis, and reshaping the tumor immune microenvironment. In recent years, immunotherapies for tumors, such as immune checkpoint inhibitors and adoptive cell immunotherapy, have been increasingly applied in clinical settings, achieving favorable outcomes. Targeting m6A modifications to modulate the immune system, such as using small-molecule inhibitors to target dysregulated m6A regulatory factors or inducing immune cell reprogramming, can enhance anti-tumor immune responses and strengthen immune cell recognition and cytotoxicity against tumor cells. m6A modification represents a new direction in tumor immunotherapy with promising clinical potential. This review discusses the regulatory role of m6A methylation on immune cells and tumor immune responses and explores new strategies for immunotherapy.
Collapse
Affiliation(s)
- Xi Chen
- Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650223, China
- NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, Yunnan, 650500, China
| | - Yixiao Yuan
- Department of Medicine, UF Health Cancer Center, University of Florida, Gainesville, FL, 32610, USA
| | - Fan Zhou
- Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650223, China
- NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, Yunnan, 650500, China
| | - Lihua Li
- NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, Yunnan, 650500, China
| | - Jun Pu
- Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650223, China.
- NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, Yunnan, 650500, China.
| | - Xiulin Jiang
- Department of Medicine, UF Health Cancer Center, University of Florida, Gainesville, FL, 32610, USA.
| |
Collapse
|
17
|
Shi T, Zhang H, Chen Y. The m6A revolution: transforming tumor immunity and enhancing immunotherapy outcomes. Cell Biosci 2025; 15:27. [PMID: 39987091 PMCID: PMC11846233 DOI: 10.1186/s13578-025-01368-z] [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: 12/09/2024] [Accepted: 02/14/2025] [Indexed: 02/24/2025] Open
Abstract
N6-methyladenosine (m6A), the most prevalent RNA modification in eukaryotes, plays a critical role in the development and progression of various diseases, including cancer, through its regulation of RNA degradation, stabilization, splicing, and cap-independent translation. Emerging evidence underscores the significant role of m6A modifications in both pro-tumorigenic and anti-tumorigenic immune responses. In this review, we provide a comprehensive overview of m6A modifications and examine the relationship between m6A regulators and cancer immune responses. Additionally, we summarize recent advances in understanding how m6A modifications influence tumor immune responses by directly modulating immune cells (e.g., dendritic cells, tumor-associated macrophages, and T cells) and indirectly affecting cancer cells via mechanisms such as cytokine and chemokine regulation, modulation of cell surface molecules, and metabolic reprogramming. Furthermore, we explore the potential synergistic effects of targeting m6A regulators in combination with immune checkpoint inhibitor (ICI) therapies. Together, this review consolidates current knowledge on the role of m6A-mediated regulation in tumor immunity, offering insights into how a deeper understanding of these modifications may identify patients who are most likely to benefit from immunotherapies.
Collapse
Affiliation(s)
- Tongguo Shi
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, 178 East Ganjiang Road, Suzhou, 215000, China.
| | - Huan Zhang
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, 178 East Ganjiang Road, Suzhou, 215000, China
| | - Yueqiu Chen
- Department of Cardiovascular Surgery of the First Affiliated Hospital and Institute for Cardiovascular Science, Suzhou Medical College of Soochow University, Soochow University, 178 East Ganjiang Road, Suzhou, 215000, China.
| |
Collapse
|
18
|
Naei VY, Tubelleza R, Monkman J, Sadeghirad H, Donovan ML, Blick T, Wicher A, Bodbin S, Viratham A, Stad R, Basu S, Cooper C, Barnett C, O'Byrne K, Ladwa R, Warkiani ME, Hughes BGM, Kulasinghe A. Spatial interaction mapping of PD-1/PD-L1 in head and neck cancer reveals the role of macrophage-tumour barriers associated with immunotherapy response. J Transl Med 2025; 23:177. [PMID: 39939997 PMCID: PMC11818323 DOI: 10.1186/s12967-025-06186-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: 10/22/2024] [Accepted: 01/29/2025] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND Mucosal head and neck squamous cell carcinoma (HNSCC) is often diagnosed at an advanced stage, where the prognosis is poor due to the high rates of recurrence and metastasis. With approximately one million new cases projected in 2024, worldwide mortality of HNSCC is estimated to reach 50% of detected cases the same year. Patients with early-stage tumours showed a 50-60% five-year survival rate in the US. Immune checkpoint inhibitors (ICIs) have shown promising results in prolonging survival in a subset of patients with recurrent or metastatic disease. However, challenges remain, particularly the limited efficacy of PD-1/PD-L1 blockade therapies. PD-L1 protein expression has been shown to be limited in its predictive power for ICI therapies. Emerging evidence shows that intricate characterisation of the tumour microenvironment (TME) is fundamental to understand interacting cells. This study aims to bridge the gap in understanding the tumor microenvironment by identifying distinct spatial patterns of PD-1/PD-L1 interactions and their association with immunotherapy responses in head and neck squamous cell carcinoma (HNSCC). METHODS In this study, we sought to apply a more nuanced approach to understanding cellular interactions by mapping PD-1/PD-L1 interactions across whole-slide HNSCC tissue samples collected prior to ICI therapy. We used a combination of spatial proteomics (Akoya Biosciences) and an in situ proximity ligation assay (isPLA, Navinci Diagnostics) to visualise PD-1/PD-L1 interactions across cell types and cellular neighbourhoods within the tumour TME. RESULTS Our findings indicate the existence of isPLA+ PD-1/PD-L1 interactions between macrophages/CD3 T cell-enriched neighbourhoods and tumour cells at the tumour-stroma boundaries in ICI-resistant tumours. The presence of these dense macrophage-tumour layers, which are either absent or dispersed in responders, indicates a barrier that may restrict immune cell infiltration and promote immune escape mechanisms. In contrast, responders had abundant B and T cell aggregates, predominantly around the tumour edges linked to enhanced immune responses to ICI therapy and better clinical outcomes. CONCLUSION This study highlights the utility of isPLA in detecting distinct tumour-immune interactions within the TME, offering new cellular interaction metrics for stratifying and optimising immunotherapy strategies.
Collapse
Affiliation(s)
- Vahid Yaghoubi Naei
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, Australia
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Rafael Tubelleza
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
- Queensland Spatial Biology Centre, Wesley Research Institute, The Wesley Hospital, Brisbane, Australia
| | - James Monkman
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
- Queensland Spatial Biology Centre, Wesley Research Institute, The Wesley Hospital, Brisbane, Australia
| | - Habib Sadeghirad
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Meg L Donovan
- Queensland Spatial Biology Centre, Wesley Research Institute, The Wesley Hospital, Brisbane, Australia
| | - Tony Blick
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | | | | | | | | | | | | | | | - Ken O'Byrne
- The Princess Alexandra Hospital, Brisbane, Australia
| | - Rahul Ladwa
- The Princess Alexandra Hospital, Brisbane, Australia
| | | | - Brett G M Hughes
- The Royal Brisbane and Women's Hospital, Brisbane, Australia
- School of Medicine, University of Queensland, Brisbane, Australia
| | - Arutha Kulasinghe
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia.
- Queensland Spatial Biology Centre, Wesley Research Institute, The Wesley Hospital, Brisbane, Australia.
| |
Collapse
|
19
|
Xie J, Du X, Li Y, Wu C, Li R, Zhao M, Shi S. Berberine shaping the tumor immune landscape via pyroptosis. Cell Immunol 2025; 408:104908. [PMID: 39701005 DOI: 10.1016/j.cellimm.2024.104908] [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: 08/04/2024] [Revised: 11/18/2024] [Accepted: 12/06/2024] [Indexed: 12/21/2024]
Abstract
Pyroptosis is a programmed cell death (PCD) mainly mediated by the Gasdermin family of proteins, among which Gasdermin E (GSDME) is considered a tumor suppressor gene. GSDME can recruit immune cells to the tumor microenvironment (TME) and promote their effects. Activating and enhancing adaptive immunity through GSDME is a potential solution for anti-tumor therapy. Here we reported that berberine (BBR), a small molecule from traditional Chinese medicine, as a GSDME activator, induced caspase-3 (C-3)/GSDME pathway-mediated pyroptosis through the mitochondrial pathway, improved the immunosuppressive state of the tumor microenvironment, and thus promoted anti-tumor immunity. We determined the induction of pyroptosis of 4 T1 cells by BBR through various experiments, and investigated the immune activation effect of BBR by co-culture in vitro, which induced DCs maturation and macrophage polarization. Zebrafish embryo toxicity experiments were used to evaluate the in vivo safety of berberine. Furthermore, the in vivo antitumor and immune activation effects of BBR were investigated using 4 T1 orthotopic model mice, and the results showed that BBR could eliminate orthotopic tumor cells by activating local and systemic immunity. Moreover, we observed that BBR significantly inhibited breast cancer lung metastasis. In summary, our results showd the role of BBR as a GSDME activator stimulated both local and systemic antitumor immune responses by inducing pyroptosis, effectively preventing tumor development and metastasis.
Collapse
Affiliation(s)
- Jinjin Xie
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xin Du
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuke Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chengyu Wu
- Department of Pharmacy, Shenzhen Technology University, Shenzhen, China.
| | - Rui Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Mengnan Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Sanjun Shi
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
20
|
Li M, Tian H, Zhuang Y, Zhang Z. New insights into N6-methyladenosine in hepatocellular carcinoma immunotherapy. Front Immunol 2025; 16:1533940. [PMID: 39911396 PMCID: PMC11794227 DOI: 10.3389/fimmu.2025.1533940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 01/03/2025] [Indexed: 02/07/2025] Open
Abstract
N6-methylation is a modification in which a methyl group is added to the adenine base of a nucleotide. This modification is crucial for controlling important functions that are vital for gene expression, including mRNA splicing, stability, and translation. Due to its intricate participation in both normal cellular processes and the course of disease, as well as its critical role in determining cell fate, N6-methyladenosine (m6A) alteration has recently attracted a lot of interest. The formation and progression of many diseases, especially cancer, can be attributed to dysregulated m6A alteration, which can cause disturbances in a variety of cellular functions, such as immunological responses, cell proliferation, and differentiation. In this study, we examine how m6A dysregulation affects hepatocellular carcinoma (HCC), with a particular emphasis on how it contributes to immunological evasion and carcinogenesis. We also investigate its potential as a novel therapeutic target, providing new perspectives on potential therapeutic approaches meant to enhance clinical results for patients with HCC.
Collapse
Affiliation(s)
- Mengran Li
- Department of Science and Technology, Taizhou Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Taizhou, Jiangsu, China
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Hu Tian
- Department of Science and Technology, Taizhou Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Taizhou, Jiangsu, China
| | - Yanshuang Zhuang
- Department of Science and Technology, Taizhou Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Taizhou, Jiangsu, China
| | - Zili Zhang
- Department of Science and Technology, Taizhou Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Taizhou, Jiangsu, China
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
21
|
Cui X, Chang M, Wang Y, Liu J, Sun Z, Sun Q, Sun Y, Ren J, Li W. Helicobacter pylori reduces METTL14-mediated VAMP3 m 6A modification and promotes the development of gastric cancer by regulating LC3C-mediated c-Met recycling. Cell Death Discov 2025; 11:13. [PMID: 39827141 PMCID: PMC11742886 DOI: 10.1038/s41420-025-02289-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 12/12/2024] [Accepted: 01/07/2025] [Indexed: 01/22/2025] Open
Abstract
Helicobacter pylori (H. pylori) plays an important role in the malignant transformation of the gastric mucosa from chronic inflammation to cancer. However, the mechanisms underlying the epigenetic regulation of gastric carcinogenesis mediated by H. pylori remain unclear. Here, we uncover that H. pylori inhibits METTL14 by upregulating ATF3. METTL14 inhibits gastric cancer (GC) cell proliferation and metastasis in vitro and in vivo. Downregulation of METTL14 inhibits Vesicle-associated membrane protein-3 (VAMP3) by reducing the m6A modification level of VAMP3 mRNA and the stability of IGF2BP2-dependent mRNA. H. pylori also accelerates the malignant progression of GC by regulating VAMP3/LC3C-mediated c-Met recycling. Moreover, the expression of METTL14 and VAMP3 in Hp+ chronic gastritis tissues is much lower than that in Hp- chronic gastritis tissues. METTL14 and VAMP3 expression levels are downregulated notably in cancerous tissues of patients with GC. Therefore, our results show a novel METTL14-VAMP3-LC3C-c-Met signalling axis in the GC development mediated by H. pylori infection, which reveals a novel m6A epigenetic modification mechanism for GC and provides potential prognostic biomarkers for GC progression.
Collapse
Affiliation(s)
- Xixi Cui
- Key Laboratory for Experimental Teratology of Chinese Ministry of Education, The Shandong Provincial Key Laboratory of Infection and Immunology, Department of Pathogenic biology, School of basic medical sciences, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| | - Mingjie Chang
- Key Laboratory for Experimental Teratology of Chinese Ministry of Education, The Shandong Provincial Key Laboratory of Infection and Immunology, Department of Pathogenic biology, School of basic medical sciences, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| | - Yuqiong Wang
- Key Laboratory for Experimental Teratology of Chinese Ministry of Education, The Shandong Provincial Key Laboratory of Infection and Immunology, Department of Pathogenic biology, School of basic medical sciences, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| | - Jiayi Liu
- Key Laboratory for Experimental Teratology of Chinese Ministry of Education, The Shandong Provincial Key Laboratory of Infection and Immunology, Department of Pathogenic biology, School of basic medical sciences, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| | - Zenghui Sun
- Key Laboratory for Experimental Teratology of Chinese Ministry of Education, The Shandong Provincial Key Laboratory of Infection and Immunology, Department of Pathogenic biology, School of basic medical sciences, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| | - Qiyu Sun
- Key Laboratory for Experimental Teratology of Chinese Ministry of Education, The Shandong Provincial Key Laboratory of Infection and Immunology, Department of Pathogenic biology, School of basic medical sciences, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| | - Yundong Sun
- Key Laboratory for Experimental Teratology of Chinese Ministry of Education, The Shandong Provincial Key Laboratory of Infection and Immunology, Department of Pathogenic biology, School of basic medical sciences, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| | - Juchao Ren
- Department of Urology, Qilu Hospital, Shandong University, Jinan, PR China
| | - Wenjuan Li
- Key Laboratory for Experimental Teratology of Chinese Ministry of Education, The Shandong Provincial Key Laboratory of Infection and Immunology, Department of Pathogenic biology, School of basic medical sciences, Cheeloo College of Medicine, Shandong University, Jinan, PR China.
| |
Collapse
|
22
|
Cesano A, Augustin R, Barrea L, Bedognetti D, Bruno TC, Carturan A, Hammer C, Ho WS, Kather JN, Kirchhoff T, Lu RO, McQuade J, Najjar YG, Pietrobon V, Ruella M, Shen R, Soldati L, Spencer C, Betof Warner A, Warren S, Ziv E, Marincola FM. Advances in the understanding and therapeutic manipulation of cancer immune responsiveness: a Society for Immunotherapy of Cancer (SITC) review. J Immunother Cancer 2025; 13:e008876. [PMID: 39824527 PMCID: PMC11749597 DOI: 10.1136/jitc-2024-008876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 12/12/2024] [Indexed: 01/20/2025] Open
Abstract
Cancer immunotherapy-including immune checkpoint inhibition (ICI) and adoptive cell therapy (ACT)-has become a standard, potentially curative treatment for a subset of advanced solid and liquid tumors. However, most patients with cancer do not benefit from the rapidly evolving improvements in the understanding of principal mechanisms determining cancer immune responsiveness (CIR); including patient-specific genetically determined and acquired factors, as well as intrinsic cancer cell biology. Though CIR is multifactorial, fundamental concepts are emerging that should be considered for the design of novel therapeutic strategies and related clinical studies. Recent advancements as well as novel approaches to address the limitations of current treatments are discussed here, with a specific focus on ICI and ACT.
Collapse
Affiliation(s)
| | - Ryan Augustin
- University of Pittsburgh Department of Medicine, Pittsburgh, Pennsylvania, USA
- Mayo Clinic, Rochester, Minnesota, USA
| | | | | | - Tullia C Bruno
- University of Pittsburgh, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | | | | | - Winson S Ho
- University of California San Francisco, San Francisco, California, USA
| | - Jakob Nikolas Kather
- Else Kroener Fresenius Center for Digital Health, Technical University Dresden, Dresden, Germany
| | - Tomas Kirchhoff
- Laura and Isaac Perlmutter Cancer Center, New York University Grossman School of Medicine, New York University Langone Health, New York, NY, USA
| | - Rongze O Lu
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California, USA
| | - Jennifer McQuade
- University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Yana G Najjar
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | | | - Marco Ruella
- University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Rhine Shen
- Kite Pharma Inc, Santa Monica, California, USA
| | | | - Christine Spencer
- Parker Institute for Cancer Immunotherapy, San Francisco, California, USA
| | | | | | - Elad Ziv
- University of California San Francisco, San Francisco, California, USA
| | | |
Collapse
|
23
|
Hayama T, Ochiai H, Ozawa T, Miyata T, Asako K, Fukushima Y, Kaneko K, Nozawa K, Fujii S, Misawa T, Fukagawa T. High systemic inflammation response index (SIRI) level as a prognostic factor for colorectal cancer patients after curative surgery: a single-center retrospective analysis. Sci Rep 2025; 15:1008. [PMID: 39762444 PMCID: PMC11704263 DOI: 10.1038/s41598-024-84991-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 12/30/2024] [Indexed: 01/11/2025] Open
Abstract
The Systemic Inflammation Response Index (SIRI), a marker used to assess systemic inflammation, is associated with lower patient survival rates in various cancer types. Factors contributing to the recurrence of colorectal cancer (CRC) have been examined previously using the preoperative SIRI. Herein, we investigated the association between the preoperative SIRI level and both the recurrence-free survival (RFS) and overall survival (OS) in patients diagnosed with CRC. We retrospectively analyzed the case of 406 patients who underwent curative surgery for Stage I-III CRC at a single institution during 2012- 2017. Based on their SIRI levels, we categorized the patients into a low-SIRI group (≤ 1700) and a high-SIRI group (> 1700). Multivariable analyses revealed that a high-SIRI level was an independent risk factor for 5-year RFS (p = 0.045) and OS (p = 0.048) in CRC patients. A Kaplan-Meier analysis demonstrated significantly poorer 5-year RFS and OS outcomes in the high-SIRI group compared to the low-SIRI group (p = 0.0001, p = 0.017 respectively). These findings suggest that the high-SIRI level is significantly associated with a poorer prognosis in patients diagnosed with CRC.
Collapse
Affiliation(s)
- Tamuro Hayama
- Department of Surgery, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi-Ku, Tokyo, 173-8605, Japan.
| | - Hiroki Ochiai
- Department of Surgery, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi-Ku, Tokyo, 173-8605, Japan
| | - Tsuyoshi Ozawa
- Department of Surgical Oncology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Toshiya Miyata
- Department of Surgery, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi-Ku, Tokyo, 173-8605, Japan
| | - Kentaro Asako
- Department of Surgery, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi-Ku, Tokyo, 173-8605, Japan
| | - Yoshihisa Fukushima
- Department of Surgery, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi-Ku, Tokyo, 173-8605, Japan
| | - Kensuke Kaneko
- Department of Surgery, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi-Ku, Tokyo, 173-8605, Japan
| | - Keijiro Nozawa
- Department of Surgery, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi-Ku, Tokyo, 173-8605, Japan
| | - Shoichi Fujii
- Department of Surgery, Yokohama General Hospital, Kanagawa, Japan
| | - Takeyuki Misawa
- Department of Surgery, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi-Ku, Tokyo, 173-8605, Japan
| | - Takeo Fukagawa
- Department of Surgery, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi-Ku, Tokyo, 173-8605, Japan
| |
Collapse
|
24
|
Wu H, Chen S, Li X, Li Y, Shi H, Qing Y, Shi B, Tang Y, Yan Z, Hao Y, Wang D, Liu W. RNA modifications in cancer. MedComm (Beijing) 2025; 6:e70042. [PMID: 39802639 PMCID: PMC11718328 DOI: 10.1002/mco2.70042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 11/12/2024] [Accepted: 11/14/2024] [Indexed: 01/16/2025] Open
Abstract
RNA modifications are emerging as critical cancer regulators that influence tumorigenesis and progression. Key modifications, such as N6-methyladenosine (m6A) and 5-methylcytosine (m5C), are implicated in various cellular processes. These modifications are regulated by proteins that write, erase, and read RNA and modulate RNA stability, splicing, translation, and degradation. Recent studies have highlighted their roles in metabolic reprogramming, signaling pathways, and cell cycle control, which are essential for tumor proliferation and survival. Despite these scientific advances, the precise mechanisms by which RNA modifications affect cancer remain inadequately understood. This review comprehensively examines the role RNA modifications play in cancer proliferation, metastasis, and programmed cell death, including apoptosis, autophagy, and ferroptosis. It explores their effects on epithelial-mesenchymal transition (EMT) and the immune microenvironment, particularly in cancer metastasis. Furthermore, RNA modifications' potential in cancer therapies, including conventional treatments, immunotherapy, and targeted therapies, is discussed. By addressing these aspects, this review aims to bridge current research gaps and underscore the therapeutic potential of targeting RNA modifications to improve cancer treatment strategies and patient outcomes.
Collapse
Affiliation(s)
- Han Wu
- Department of Oral and Maxillofacial SurgeryHospital of StomatologyJilin University, ChangchunJilin provinceChina
- Jilin Provincial Key Laboratory of Tooth Development and Bone RemodelingHospital of StomatologyJilin University, ChangchunJilin provincleChina
| | - Shi Chen
- Department of Oral and Maxillofacial SurgeryHospital of StomatologyJilin University, ChangchunJilin provinceChina
- Jilin Provincial Key Laboratory of Tooth Development and Bone RemodelingHospital of StomatologyJilin University, ChangchunJilin provincleChina
| | - Xiang Li
- Department of Oral and Maxillofacial SurgeryHospital of StomatologyJilin University, ChangchunJilin provinceChina
- Jilin Provincial Key Laboratory of Tooth Development and Bone RemodelingHospital of StomatologyJilin University, ChangchunJilin provincleChina
| | - Yuyang Li
- Department of Oral and Maxillofacial SurgeryHospital of StomatologyJilin University, ChangchunJilin provinceChina
- Jilin Provincial Key Laboratory of Tooth Development and Bone RemodelingHospital of StomatologyJilin University, ChangchunJilin provincleChina
| | - He Shi
- Department of Oral and Maxillofacial SurgeryHospital of StomatologyJilin University, ChangchunJilin provinceChina
- Jilin Provincial Key Laboratory of Tooth Development and Bone RemodelingHospital of StomatologyJilin University, ChangchunJilin provincleChina
| | - Yiwen Qing
- Department of Oral and Maxillofacial SurgeryHospital of StomatologyJilin University, ChangchunJilin provinceChina
- Jilin Provincial Key Laboratory of Tooth Development and Bone RemodelingHospital of StomatologyJilin University, ChangchunJilin provincleChina
| | - Bohe Shi
- Laboratory Animal CenterCollege of Animal ScienceJilin University, ChangchunJilin provinceChina
| | - Yifei Tang
- Laboratory Animal CenterCollege of Animal ScienceJilin University, ChangchunJilin provinceChina
| | - Zhuoyi Yan
- Laboratory Animal CenterCollege of Animal ScienceJilin University, ChangchunJilin provinceChina
| | - Yang Hao
- Laboratory Animal CenterCollege of Animal ScienceJilin University, ChangchunJilin provinceChina
| | - Dongxu Wang
- Laboratory Animal CenterCollege of Animal ScienceJilin University, ChangchunJilin provinceChina
| | - Weiwei Liu
- Department of Oral and Maxillofacial SurgeryHospital of StomatologyJilin University, ChangchunJilin provinceChina
- Jilin Provincial Key Laboratory of Tooth Development and Bone RemodelingHospital of StomatologyJilin University, ChangchunJilin provincleChina
| |
Collapse
|
25
|
Liu J, Lu J, Wu L, Zhang T, Wu J, Li L, Tai Z, Chen Z, Zhu Q. Targeting tumor-associated macrophages: Novel insights into immunotherapy of skin cancer. J Adv Res 2025; 67:231-252. [PMID: 38242529 PMCID: PMC11725115 DOI: 10.1016/j.jare.2024.01.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 12/19/2023] [Accepted: 01/11/2024] [Indexed: 01/21/2024] Open
Abstract
BACKGROUND The incidence of skin cancer is currently increasing, and conventional treatment options inadequately address the demands of disease management. Fortunately, the recent rapid advancement of immunotherapy, particularly immune checkpoint inhibitors (ICIs), has ushered in a new era for numerous cancer patients. However, the efficacy of immunotherapy remains suboptimal due to the impact of the tumor microenvironment (TME). Tumor-associated macrophages (TAMs), a major component of the TME, play crucial roles in tumor invasion, metastasis, angiogenesis, and immune evasion, significantly impacting tumor development. Consequently, TAMs have gained considerable attention in recent years, and their roles have been extensively studied in various tumors. However, the specific roles of TAMs and their regulatory mechanisms in skin cancer remain unclear. AIM OF REVIEW This paper aims to elucidate the origin and classification of TAMs, investigate the interactions between TAMs and various immune cells, comprehensively understand the precise mechanisms by which TAMs contribute to the pathogenesis of different types of skin cancer, and finally discuss current strategies for targeting TAMs in the treatment of skin cancer. KEY SCIENTIFIC CONCEPTS OF OVERVIEW With a specific emphasis on the interrelationship between TAMs and skin cancer, this paper posits that therapeutic modalities centered on TAMs hold promise in augmenting and harmonizing with prevailing clinical interventions for skin cancer, thereby charting a novel trajectory for advancing the landscape of immunotherapeutic approaches for skin cancer.
Collapse
Affiliation(s)
- Jun Liu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, 1278 Baode Road, Shanghai 200443, China; Shanghai Engineering Research Center of Topical Chinese Medicine, 1278 Baode Road, Shanghai 200443, China
| | - Jiaye Lu
- School of Medicine, Shanghai University, 99 Shangda Road, Shanghai 200444, China; Shanghai Engineering Research Center of Topical Chinese Medicine, 1278 Baode Road, Shanghai 200443, China
| | - Ling Wu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, 1278 Baode Road, Shanghai 200443, China; Shanghai Engineering Research Center of Topical Chinese Medicine, 1278 Baode Road, Shanghai 200443, China
| | - Tingrui Zhang
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, 1278 Baode Road, Shanghai 200443, China; Shanghai Engineering Research Center of Topical Chinese Medicine, 1278 Baode Road, Shanghai 200443, China
| | - Junchao Wu
- School of Medicine, Shanghai University, 99 Shangda Road, Shanghai 200444, China; Shanghai Engineering Research Center of Topical Chinese Medicine, 1278 Baode Road, Shanghai 200443, China
| | - Lisha Li
- School of Medicine, Shanghai University, 99 Shangda Road, Shanghai 200444, China; Shanghai Engineering Research Center of Topical Chinese Medicine, 1278 Baode Road, Shanghai 200443, China
| | - Zongguang Tai
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, 1278 Baode Road, Shanghai 200443, China; Shanghai Engineering Research Center of Topical Chinese Medicine, 1278 Baode Road, Shanghai 200443, China.
| | - Zhongjian Chen
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, 1278 Baode Road, Shanghai 200443, China; Shanghai Engineering Research Center of Topical Chinese Medicine, 1278 Baode Road, Shanghai 200443, China.
| | - Quangang Zhu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, 1278 Baode Road, Shanghai 200443, China; Shanghai Engineering Research Center of Topical Chinese Medicine, 1278 Baode Road, Shanghai 200443, China.
| |
Collapse
|
26
|
Li X, Peng L, Yang X, Luo J, Wang J, Mou K, Zhou H, Luo Y, Xiang L. N6-methyladenosine RNA methylation, a new hallmark of metabolic reprogramming in the immune microenvironment. Front Immunol 2024; 15:1464042. [PMID: 39759516 PMCID: PMC11695279 DOI: 10.3389/fimmu.2024.1464042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 12/09/2024] [Indexed: 01/07/2025] Open
Abstract
N6-methyladenosine is one of the most common and reversible post-transcriptional modifications in eukaryotes, and it is involved in alternative splicing and RNA transcription, degradation, and translation. It is well known that cancer cells acquire energy through metabolic reprogramming to exhibit various biological behaviors. Moreover, numerous studies have demonstrated that m6A induces cancer metabolic reprogramming by regulating the expression of core metabolic genes or by activating metabolic signaling pathways. Meanwhile, m6A modifications and related regulators are key targets in the regulation of immune effects. We further summarize how m6A modifications contribute to tumor metabolism, and how these events affect the tumor immune microenvironment, with a specific focus on different cell types. Finally, we focus on the specific applications of this field to tumor immunotherapy. We review the potential role of m6A in metabolic reprogramming of tumor immune microenvironment and its regulatory mechanism, with the aim of providing new targets for tumor metabolic regulation and immunotherapy.
Collapse
Affiliation(s)
- Xiaoyue Li
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- School of Life Sciences, Yunnan University, Kunming, China
| | - Lin Peng
- Department of Bone and Joint, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xuelian Yang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jing Luo
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jianmei Wang
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Kelin Mou
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Huan Zhou
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yuhao Luo
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Li Xiang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
27
|
Huart C, Gupta MS, Van Ginderachter JA. The role of RNA modifications in disease-associated macrophages. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102315. [PMID: 39296330 PMCID: PMC11408368 DOI: 10.1016/j.omtn.2024.102315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/21/2024]
Abstract
In recent years, the field of epitranscriptomics has witnessed significant breakthroughs with the identification of more than 150 different chemical modifications in different RNA species. It has become increasingly clear that these chemical modifications play an important role in the regulation of fundamental processes linked to cell fate and development. Further interest was sparked by the ability of the epitranscriptome to regulate pathogenesis. However, despite the involvement of macrophages in a multitude of diseases, a clear knowledge gap exists in the understanding of how RNA modifications regulate the phenotype of these cells. Here, we provide a comprehensive overview of the known roles of macrophage RNA modifications in the context of different diseases.
Collapse
Affiliation(s)
- Camille Huart
- Lab of Cellular and Molecular Immunology, Brussels Center for Immunology (BCIM), Vrije Universiteit Brussel, Brussels, Belgium
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
| | - Mayuk Saibal Gupta
- Lab of Cellular and Molecular Immunology, Brussels Center for Immunology (BCIM), Vrije Universiteit Brussel, Brussels, Belgium
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
| | - Jo A Van Ginderachter
- Lab of Cellular and Molecular Immunology, Brussels Center for Immunology (BCIM), Vrije Universiteit Brussel, Brussels, Belgium
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
| |
Collapse
|
28
|
Yoon J, Fagan E, Jeong M, Park JH. In Situ Tumor-Infiltrating Lymphocyte Therapy by Local Delivery of an mRNA Encoding Membrane-Anchored Anti-CD3 Single-Chain Variable Fragment. ACS NANO 2024; 18:32401-32420. [PMID: 39527145 DOI: 10.1021/acsnano.4c03518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Tumor-infiltrating lymphocyte (TIL) therapy has shown promising responses in clinical trials for highly aggressive cancers such as advanced melanoma and metastatic colorectal cancer. However, TIL therapy is still limited in clinical practice due to the complex ex vivo cell preparation process. Here, we report an "in situ TIL therapy" for the treatment of solid tumors. We utilized lipid nanoparticles for the delivery of an mRNA encoding membrane-anchored anti-CD3 single-chain variable fragment (scFv) (MA-aCD3), efficiently engineering both tumor-associated macrophages (TAMs) and tumor cells following intratumoral delivery. Expression of MA-aCD3 resulted in enhanced TIL activation, proliferation, and tumor cell engagement directly within the tumor microenvironment. In B16F10 and MC38 tumor models, concurrent expression of MA-aCD3 on TAMs and tumor cells mediated by mRNA delivery resulted in significant antitumor effects via in situ polyclonal CD8+ TIL expansion and directed cytotoxic effector functions. In addition, combinatorial treatment of MA-aCD3-encoding mRNA and antiprogrammed cell death 1 (anti-PD-1) antibodies exhibited synergistic antitumor effects on anti-PD-1 refractory B16F10 tumors. Together, our findings suggest that in situ TIL therapy is a practical and effective mRNA-based therapeutic modality for the treatment of solid tumors.
Collapse
Affiliation(s)
- Junyong Yoon
- Department of Bio and Brain Engineering and KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology (KAIST), Yuseong-gu, Daejeon34141, Republic of Korea
| | - Erinn Fagan
- Department of Bio and Brain Engineering and KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology (KAIST), Yuseong-gu, Daejeon34141, Republic of Korea
| | - Moonkyoung Jeong
- Department of Bio and Brain Engineering and KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology (KAIST), Yuseong-gu, Daejeon34141, Republic of Korea
| | - Ji-Ho Park
- Department of Bio and Brain Engineering and KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology (KAIST), Yuseong-gu, Daejeon34141, Republic of Korea
| |
Collapse
|
29
|
Li J, Wang P, Liu F, Li Y, Wu Y, Wang F, Du J. Methyltransferase-like 14 promotes the tumorigenesis and proliferation of pancreatic cancer cells through myc proto-oncogene signaling pathway. Cytojournal 2024; 21:55. [PMID: 39737122 PMCID: PMC11683408 DOI: 10.25259/cytojournal_105_2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 09/23/2024] [Indexed: 01/01/2025] Open
Abstract
Objective Pancreatic cancer is characterized by low survival rate and rapid deterioration. Methyltransferase-like 14 (METTL14), as N6-methyladenosine (m6A) methyltransferase, is closely related to tumor progression. The purpose of this study is to look into how METTL14 affects pancreatic cancer tumorigenesis, cell division, and apoptosis. Material and Methods We examined and contrasted the levels of METTL14 protein and messenger RNA expression in human pancreatic ductal cells and human pancreatic cancer cells. After silencing or upregulating METTL14, the proliferative ability, migration ability, and cell apoptosis of pancreatic tumor cells was detected by colony-forming assay, wound scratch healing assay, cell counting kit 8 assay, and terminal deoxynucleotidyl transferasemediated 2'-deoxyuridine 5'-triphosphate-biotin nick end labeling assay. Following the use of c-Myc inhibitor (10058-F4), western blot analysis was carried out to investigate the key factor expression and c-Myc signaling pathway activation status. Results METTL14 was preferentially expressed in human pancreatic cancer cells PANC-1 and SW1990 than in human normal pancreatic duct cells human pancreatic nestin-expressing cells (HPNE) (P < 0.001). Overexpression of METTL14 increased the tumorigenic and proliferative ability of pancreatic cancer cells. Overexpression of METTL14 decreased apoptosis rate. Western blot assay showed that nucleus b-catenin increased when METTL14 was overexpressed, and nucleus b-catenin decreased when METTL14 was silenced in PANC-1 cell (P < 0.01). The protein expression of other key factors, such as c-Myc, matrix metalloproteinase (MMP)-9, and MMP-2, were also affected. The use of c-Myc inhibitor (10058-F4) on the basis of OE-METTL14 reversed the effect of the overexpression of METTL14 on promoting the tumorigenesis and cell proliferation of pancreatic cancer cell lines PANC-1 and SW1990. Conclusion METTL14 promoted the tumorigenesis and proliferation of pancreatic cancer cells by the c-Myc signaling pathway.
Collapse
Affiliation(s)
- Junru Li
- Department of General Surgery, Jincheng General Hospital, Jincheng, China
| | - Peng Wang
- Department of General Surgery, Jincheng General Hospital, Jincheng, China
| | - Fei Liu
- Department of General Surgery, Jincheng General Hospital, Jincheng, China
| | - Yuanyuan Li
- Department of General Surgery, Jincheng General Hospital, Jincheng, China
| | - Youyou Wu
- Department of General Surgery, Jincheng General Hospital, Jincheng, China
| | - Fengbo Wang
- Department of General Surgery, Jincheng General Hospital, Jincheng, China
| | - Jundong Du
- Department of General Surgery, Jincheng General Hospital, Jincheng, China
| |
Collapse
|
30
|
Tang Y, Tang S, Yang W, Zhang Z, Wang T, Wu Y, Xu J, Pilarsky C, Mazzone M, Wang LW, Sun Y, Tian R, Tang Y, Wang Y, Wang C, Xue J. MED12 loss activates endogenous retroelements to sensitise immunotherapy in pancreatic cancer. Gut 2024; 73:1999-2011. [PMID: 39216984 DOI: 10.1136/gutjnl-2024-332350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Abstract
OBJECTIVE Pancreatic ductal adenocarcinoma (PDAC) stands as one of the most lethal cancers, marked by its lethality and limited treatment options, including the utilisation of checkpoint blockade (ICB) immunotherapy. Epigenetic dysregulation is a defining feature of tumourigenesis that is implicated in immune surveillance, but remains elusive in PDAC. DESIGN To identify the factors that modulate immune surveillance, we employed in vivo epigenetic-focused CRISPR-Cas9 screen in mouse PDAC tumour models engrafted in either immunocompetent or immunodeficient mice. RESULTS Here, we identified MED12 as a top hit, emerging as a potent negative modulator of immune tumour microenviroment (TME) in PDAC. Loss of Med12 significantly promoted infiltration and cytotoxicity of immune cells including CD8+ T cells, natural killer (NK) and NK1.1+ T cells in tumours, thereby heightening the sensitivity of ICB treatment in a mouse model of PDAC. Mechanistically, MED12 stabilised heterochromatin protein HP1A to repress H3K9me3-marked endogenous retroelements. The derepression of retrotransposons induced by MED12 loss triggered cytosolic nucleic acid sensing and subsequent activation of type I interferon pathways, ultimately leading to robust inflamed TME . Moreover, we uncovered a negative correlation between MED12 expression and immune resposne pathways, retrotransposon levels as well as the prognosis of patients with PDAC undergoing ICB therapy. CONCLUSION In summary, our findings underscore the pivotal role of MED12 in remodelling immnue TME through the epigenetic silencing of retrotransposons, offering a potential therapeutic target for enhancing tumour immunogenicity and overcoming immunotherapy resistance in PDAC.
Collapse
Affiliation(s)
- Yingying Tang
- State Key Laboratory of Systems Medicine for Cancer, Stem Cell Research Center, Ren Ji Hospital, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shijie Tang
- Centre of Biomedical Systems and Informatics, ZJU-UoE Institute, Zhejiang University School of Medicine, International Campus, Zhejiang University, Haining, Zhejiang, China
| | - Wenjuan Yang
- State Key Laboratory of Systems Medicine for Cancer, Stem Cell Research Center, Ren Ji Hospital, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhengyan Zhang
- State Key Laboratory of Systems Medicine for Cancer, Stem Cell Research Center, Ren Ji Hospital, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Teng Wang
- Centre of Biomedical Systems and Informatics, ZJU-UoE Institute, Zhejiang University School of Medicine, International Campus, Zhejiang University, Haining, Zhejiang, China
| | - Yuyun Wu
- Centre of Biomedical Systems and Informatics, ZJU-UoE Institute, Zhejiang University School of Medicine, International Campus, Zhejiang University, Haining, Zhejiang, China
| | - Junyi Xu
- State Key Laboratory of Systems Medicine for Cancer, Stem Cell Research Center, Ren Ji Hospital, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Christian Pilarsky
- Department of Surgery, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Massimiliano Mazzone
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium, Leuven, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, Leuven, Belgium
| | - Lei-Wei Wang
- Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yongwei Sun
- Department of Biliary and Pancreatic Surgery, Shanghai Jiao Tong University School of Medicine Affiliated Renji Hospital, Shanghai, China
| | - Ruijun Tian
- Shenzhen Key Laboratory of Functional Proteomics, Guangming Advanced Research Institute, Southern University of Science and Technology, Shenzhen, China
| | - Yujie Tang
- Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Key Laboratory of Reproductive Medicine, Department of Histoembryology, Genetics and Developmental Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Wang
- Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Chaochen Wang
- Centre of Biomedical Systems and Informatics, ZJU-UoE Institute, Zhejiang University School of Medicine, International Campus, Zhejiang University, Haining, Zhejiang, China
- Department of Breast Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Hangzhou, China
- Biomedical and Health Translational Research Centre, Zhejiang University, Zhejiang, China
| | - Jing Xue
- State Key Laboratory of Systems Medicine for Cancer, Stem Cell Research Center, Ren Ji Hospital, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
31
|
Zhang H, Luo X, Yang W, Wu Z, Zhao Z, Pei X, Zhang X, Chen C, Lei JH, Shi Q, Zhao Q, Chen Y, Wu W, Zeng Z, Ju HQ, Qiu M, Liu J, Shen B, Chen M, Chen J, Deng CX, Xu RH, Hou J. YTHDF2 upregulation and subcellular localization dictate CD8 T cell polyfunctionality in anti-tumor immunity. Nat Commun 2024; 15:9559. [PMID: 39500904 PMCID: PMC11538425 DOI: 10.1038/s41467-024-53997-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 10/28/2024] [Indexed: 11/08/2024] Open
Abstract
RNA methylation is an important regulatory process to determine immune cell function but how it affects the anti-tumor activity of CD8 T cells is not fully understood. Here we show that the N6-methyladenosine (m6A) RNA reader YTHDF2 is highly expressed in early effector or effector-like CD8 T cells. We find that YTHDF2 facilitates nascent RNA synthesis, and m6A recognition is fundamental for this distinctively nuclear function of the protein, which also reinforces its autoregulation at the RNA level. Loss of YTHDF2 in T cells exacerbates tumor progression and confers unresponsiveness to PD-1 blockade in mice and in humans. In addition to initiating RNA decay that is necessary for mitochondrial fitness, YTHDF2 orchestrates chromatin changes that promote T cell polyfunctionality. YTHDF2 interacts with IKZF1/3, which is important for sustained transcription of their target genes. Accordingly, immunotherapy-induced efficacy could be largely restored in YTHDF2-deficient T cells through combinational use of IKZF1/3 inhibitor lenalidomide in a mouse model. Thus, YTHDF2 coordinates epi-transcriptional and transcriptional networks to potentiate T cell immunity, which could inform therapeutic intervention.
Collapse
Affiliation(s)
- Haiyan Zhang
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau SAR, China; MOE Frontier Science Center for Precision Oncology, University of Macau, Macau, SAR, China
| | - Xiaojing Luo
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, China
| | - Wei Yang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Translational Research Center, Zhuhai UM Science & Technology Research Institute, Zhuhai, China
| | - Zhiying Wu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, China
| | - Zhicong Zhao
- Department of Systems Biology, The Beckman Research Institute of City of Hope, Duarte, CA, USA
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xin Pei
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau SAR, China; MOE Frontier Science Center for Precision Oncology, University of Macau, Macau, SAR, China
| | - Xue Zhang
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau SAR, China; MOE Frontier Science Center for Precision Oncology, University of Macau, Macau, SAR, China
| | - Chonghao Chen
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau SAR, China; MOE Frontier Science Center for Precision Oncology, University of Macau, Macau, SAR, China
| | - Josh Haipeng Lei
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau SAR, China; MOE Frontier Science Center for Precision Oncology, University of Macau, Macau, SAR, China
| | - Qingxia Shi
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau SAR, China; MOE Frontier Science Center for Precision Oncology, University of Macau, Macau, SAR, China
| | - Qi Zhao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, China
| | - Yanxing Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, China
| | - Wenwei Wu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, China
| | - Zhaolei Zeng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, China
| | - Huai-Qiang Ju
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, China
| | - Miaozhen Qiu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, China
| | - Jun Liu
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Bin Shen
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Minshan Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Liver Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jianjun Chen
- Department of Systems Biology, The Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Chu-Xia Deng
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau SAR, China; MOE Frontier Science Center for Precision Oncology, University of Macau, Macau, SAR, China
- Translational Research Center, Zhuhai UM Science & Technology Research Institute, Zhuhai, China
| | - Rui-Hua Xu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China.
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, China.
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China.
| | - Jiajie Hou
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau SAR, China; MOE Frontier Science Center for Precision Oncology, University of Macau, Macau, SAR, China.
- Translational Research Center, Zhuhai UM Science & Technology Research Institute, Zhuhai, China.
- Department of Liver Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China.
| |
Collapse
|
32
|
Haag SM, Xie S, Eidenschenk C, Fortin JP, Callow M, Costa M, Lun A, Cox C, Wu SZ, Pradhan RN, Lock J, Kuhn JA, Holokai L, Thai M, Freund E, Nissenbaum A, Keir M, Bohlen CJ, Martin S, Geiger-Schuller K, Hejase HA, Yaspan BL, Melo Carlos S, Turley SJ, Murthy A. Systematic perturbation screens identify regulators of inflammatory macrophage states and a role for TNF mRNA m6A modification. Nat Genet 2024; 56:2493-2505. [PMID: 39443811 DOI: 10.1038/s41588-024-01962-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Accepted: 09/26/2024] [Indexed: 10/25/2024]
Abstract
Macrophages exhibit remarkable functional plasticity, a requirement for their central role in tissue homeostasis. During chronic inflammation, macrophages acquire sustained inflammatory 'states' that contribute to disease, but there is limited understanding of the regulatory mechanisms that drive their generation. Here we describe a systematic functional genomics approach that combines genome-wide phenotypic screening in primary murine macrophages with transcriptional and cytokine profiling of genetic perturbations in primary human macrophages to uncover regulatory circuits of inflammatory states. This process identifies regulators of five distinct states associated with key features of macrophage function. Among these regulators, loss of the N6-methyladenosine (m6A) writer components abolishes m6A modification of TNF transcripts, thereby enhancing mRNA stability and TNF production associated with multiple inflammatory pathologies. Thus, phenotypic characterization of primary murine and human macrophages describes the regulatory circuits underlying distinct inflammatory states, revealing post-transcriptional control of TNF mRNA stability as an immunosuppressive mechanism in innate immunity.
Collapse
Affiliation(s)
| | - Shiqi Xie
- Genentech Inc., South San Francisco, CA, USA
| | | | | | | | - Mike Costa
- Genentech Inc., South San Francisco, CA, USA
| | - Aaron Lun
- Genentech Inc., South San Francisco, CA, USA
| | - Chris Cox
- Genentech Inc., South San Francisco, CA, USA
| | - Sunny Z Wu
- Genentech Inc., South San Francisco, CA, USA
| | | | - Jaclyn Lock
- Genentech Inc., South San Francisco, CA, USA
- Sana Biotechnology Inc., South San Francisco, CA, USA
| | - Julia A Kuhn
- Genentech Inc., South San Francisco, CA, USA
- Alector Therapeutics, South San Francisco, CA, USA
| | | | - Minh Thai
- Genentech Inc., South San Francisco, CA, USA
| | | | | | - Mary Keir
- Genentech Inc., South San Francisco, CA, USA
| | | | | | | | | | | | | | | | - Aditya Murthy
- Genentech Inc., South San Francisco, CA, USA.
- Gilead Sciences, Foster City, CA, USA.
| |
Collapse
|
33
|
Li T, Huang Y, Cui S, Hong Z, Zhang X, Li Z, Chen K, Chen D. RNA methylation patterns of tumor microenvironment cells regulate prognosis and immunotherapeutic responsiveness in patients with triple-negative breast cancer. Sci Rep 2024; 14:26075. [PMID: 39478153 PMCID: PMC11525934 DOI: 10.1038/s41598-024-77941-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 10/28/2024] [Indexed: 11/02/2024] Open
Abstract
Immunotherapy research focuses on reshaping the tumor microenvironment (TME) to enhance its antitumor immune responses, with an emphasis on understanding the impact of RNA methylation in triple-negative breast cancer (TNBC) TME regulation. This study explored the influence of various RNA methyltransferases on TME cells in TNBC and their correlation with prognosis and immunotherapy response. Using non-negative matrix factorization on single-cell RNA-sequencing data, distinct TME cell clusters were identified based on the expression of 30 RNA methyltransferases. Various analyses, including pseudotime, cell communication, transcription factor regulatory network, and gene enrichment, were conducted on these clusters. The roles of RNA methyltransferase-mediated TME clusters in prognosis and immunotherapy response were determined using TNBC bulk RNA-Seq data, and the findings were validated through immunofluorescence analysis of a tissue microarray comprising 87 samples. Spatial transcriptomic analysis further revealed the distribution of TME cell clusters. Different methyltransferase-mediated cell clusters exhibited unique metabolic, immune, transcriptional, and intercellular communication patterns. Survival analysis indicated prognostic significance in specific TME cell clusters, and immunofluorescence analysis confirmed the prognostic value of m6A_WTAP + CD8T + cells. In conclusion, our study illustrated the involvement of these cell subgroups in tumor growth and antitumor immunity modulation, providing insights into the enhancement of TNBC immunotherapy.
Collapse
Affiliation(s)
- Tingjun Li
- The School of Clinical Medicine, Fujian Medical University, Fuzhou, China
- Department of Breast Surgery, Quanzhou First Hospital of Fujian Medical University, Quanzhou, China
| | - Yiqin Huang
- The School of Clinical Medicine, Fujian Medical University, Fuzhou, China
- Department of Breast Surgery, Quanzhou First Hospital of Fujian Medical University, Quanzhou, China
| | - Shien Cui
- Breast Center of Zhongshan City People's Hospital, Zhongshan, China
| | - Zhipeng Hong
- Department of Breast Surgery, Quanzhou First Hospital of Fujian Medical University, Quanzhou, China
| | - Xinhai Zhang
- The School of Clinical Medicine, Fujian Medical University, Fuzhou, China
- Department of Breast Surgery, Quanzhou First Hospital of Fujian Medical University, Quanzhou, China
| | - Zhihao Li
- The School of Clinical Medicine, Fujian Medical University, Fuzhou, China
- Department of Breast Surgery, Quanzhou First Hospital of Fujian Medical University, Quanzhou, China
| | - Kunqi Chen
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China.
| | - Debo Chen
- The School of Clinical Medicine, Fujian Medical University, Fuzhou, China.
- Department of Breast Surgery, Quanzhou First Hospital of Fujian Medical University, Quanzhou, China.
| |
Collapse
|
34
|
Fan Q, Fu ZW, Xu M, Lv F, Shi JS, Zeng QQ, Xiong DH. Research progress of tumor-associated macrophages in immune checkpoint inhibitor tolerance in colorectal cancer. World J Gastrointest Oncol 2024; 16:4064-4079. [DOI: 10.4251/wjgo.v16.i10.4064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 08/03/2024] [Accepted: 08/16/2024] [Indexed: 09/26/2024] Open
Abstract
The relevant mechanism of tumor-associated macrophages (TAMs) in the treatment of colorectal cancer patients with immune checkpoint inhibitors (ICIs) is discussed, and the application prospects of TAMs in reversing the treatment tolerance of ICIs are discussed to provide a reference for related studies. As a class of drugs widely used in clinical tumor immunotherapy, ICIs can act on regulatory molecules on cells that play an inhibitory role-immune checkpoints-and kill tumors in the form of an immune response by activating a variety of immune cells in the immune system. The sensitivity of patients with different types of colorectal cancer to ICI treatment varies greatly. The phenotype and function of TAMs in the colorectal cancer microenvironment are closely related to the efficacy of ICIs. ICIs can regulate the phenotypic function of TAMs, and TAMs can also affect the tolerance of colorectal cancer to ICI therapy. TAMs play an important role in ICI resistance, and making full use of this target as a therapeutic strategy is expected to improve the immunotherapy efficacy and prognosis of patients with colorectal cancer.
Collapse
Affiliation(s)
- Qi Fan
- Intestinal Center, Chongqing University Three Gorges Hospital, Chongqing 404000, China
| | - Zheng-Wei Fu
- Intestinal Center, Chongqing University Three Gorges Hospital, Chongqing 404000, China
| | - Ming Xu
- Intestinal Center, Chongqing University Three Gorges Hospital, Chongqing 404000, China
| | - Feng Lv
- Intestinal Center, Chongqing University Three Gorges Hospital, Chongqing 404000, China
| | - Jia-Song Shi
- Intestinal Center, Chongqing University Three Gorges Hospital, Chongqing 404000, China
| | - Qi-Qi Zeng
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu Province, China
| | - De-Hai Xiong
- Intestinal Center, Chongqing University Three Gorges Hospital, Chongqing 404000, China
| |
Collapse
|
35
|
Miyauchi S, Arimoto KI, Liu M, Zhang Y, Zhang DE. Protocol to study the immune profile of syngeneic mouse tumor models. STAR Protoc 2024; 5:103139. [PMID: 38878286 PMCID: PMC11234017 DOI: 10.1016/j.xpro.2024.103139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 04/29/2024] [Accepted: 05/30/2024] [Indexed: 07/13/2024] Open
Abstract
Flow cytometry, single-cell RNA sequencing, and other analyses enable us to capture immune profiles of the tumor microenvironment. Here, we present a protocol to characterize the immune profile of tumor-bearing mice. We describe steps for establishing mouse models and preparing single-cell suspensions from tumor tissue and other immune-related organs, which can be further analyzed by flow cytometry and other omics assays. We then detail procedures for staining, flow cytometry analysis, and phenotyping of the immune cell populations. For complete details on the use and execution of this protocol, please refer to Miyauchi et al.1.
Collapse
Affiliation(s)
- Sayuri Miyauchi
- Moores Cancer Center, University of California, San Diego, La Jolla, CA 92037, USA
| | - Kei-Ichiro Arimoto
- Moores Cancer Center, University of California, San Diego, La Jolla, CA 92037, USA
| | - Mengdan Liu
- Moores Cancer Center, University of California, San Diego, La Jolla, CA 92037, USA; School of Biological Sciences, University of California, San Diego, La Jolla, CA 92037, USA
| | - Yue Zhang
- Moores Cancer Center, University of California, San Diego, La Jolla, CA 92037, USA; School of Biological Sciences, University of California, San Diego, La Jolla, CA 92037, USA
| | - Dong-Er Zhang
- Moores Cancer Center, University of California, San Diego, La Jolla, CA 92037, USA; School of Biological Sciences, University of California, San Diego, La Jolla, CA 92037, USA; Department of Pathology, University of California, San Diego, La Jolla, CA 92037, USA.
| |
Collapse
|
36
|
Qin X, Liu H, Zhang Q, Che Y, Lei T, Tang F, Hu Q. RNA modifications in cancer immune therapy: regulators of immune cells and immune checkpoints. Front Immunol 2024; 15:1463847. [PMID: 39372415 PMCID: PMC11449722 DOI: 10.3389/fimmu.2024.1463847] [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: 07/12/2024] [Accepted: 09/02/2024] [Indexed: 10/08/2024] Open
Abstract
RNA modifications are epigenetic changes that alter the structure and function of RNA molecules, playing a crucial role in the onset, progression, and treatment of cancer. Immune checkpoint inhibitor (ICI) therapies, particularly PD-1 blockade and anti-CTLA-4 treatments, have changed the treatment landscape of virous cancers, showing great potential in the treatment of different cancer patients, but sensitivity to these therapies is limited to certain individuals. This review offers a comprehensive survey of the functions and therapeutic implications of the four principal RNA modifications, particularly highlighting the significance of m6A in the realms of immune cells in tumor and immunotherapy. This review starts by providing a foundational summary of the roles RNA modifications assume within the immune cell community, focusing on T cells, NK cells, macrophages, and dendritic cells. We then discuss how RNA modifications influence the intricate regulatory mechanisms governing immune checkpoint expression, modulation of ICI efficacy, and prediction of ICI treatment outcomes, and review drug therapies targeting genes regulated by RNA modifications. Finally, we explore the role of RNA modifications in gene editing, cancer vaccines, and adoptive T cell therapies, offering valuable insights into the use of RNA modifications in cancer immunotherapy.
Collapse
Affiliation(s)
- Xiangyu Qin
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, China
- Renmin Hospital of Wuhan Economic and Technological Development Zone (Hannan), Wuhan, China
- Wuhan University Heavy Ion Medicine Center, Wuhan, China
| | - Huali Liu
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qixuan Zhang
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yuhang Che
- Renmin Hospital of Wuhan Economic and Technological Development Zone (Hannan), Wuhan, China
- Wuhan University Heavy Ion Medicine Center, Wuhan, China
| | - Tianyu Lei
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, China
- Renmin Hospital of Wuhan Economic and Technological Development Zone (Hannan), Wuhan, China
- Wuhan University Heavy Ion Medicine Center, Wuhan, China
| | - Fang Tang
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Qinyong Hu
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, China
- Renmin Hospital of Wuhan Economic and Technological Development Zone (Hannan), Wuhan, China
- Wuhan University Heavy Ion Medicine Center, Wuhan, China
| |
Collapse
|
37
|
Shao Y, Han S, Hou Z, Yang C, Zhao Y. Tumor-associated macrophages within the immunological milieu: An emerging focal point for therapeutic intervention. Heliyon 2024; 10:e36839. [PMID: 39281573 PMCID: PMC11401039 DOI: 10.1016/j.heliyon.2024.e36839] [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: 02/03/2024] [Revised: 08/21/2024] [Accepted: 08/22/2024] [Indexed: 09/18/2024] Open
Abstract
Tumor-associated macrophages play an important role in the tumor immune microenvironment, and regulating the function of tumor-associated macrophages has important therapeutic potential in tumor therapy. Mature macrophages could migrate to the tumor microenvironment, influencing multiple factors such as tumor cell proliferation, invasion, metastasis, extracellular matrix remodeling, immune suppression, and drug resistance. As a major component of the tumor microenvironment, tumor-associated macrophages crosstalk with other immune cells. Currently, tumor-associated macrophages have garnered considerable attention in tumor therapy, broadening the spectrum of drug selection to some extent, thereby aiding in mitigating the prevailing clinical drug resistance dilemma. This article summarizes the recent advances in tumor-associated macrophages concerning immunology, drug targeting mechanisms for tumor-associated macrophages treatment, new developments, and existing challenges, offering insights for future therapeutic approaches. In addition, this paper summarized the impact of tumor-associated macrophages on current clinical therapies, discussed the advantages and disadvantages of targeted tumor-associated macrophages therapy compared with existing tumor therapies, and predicted and discussed the future role of targeted tumor-associated macrophages therapy and the issues that need to be focused on.
Collapse
Affiliation(s)
- Yanchi Shao
- Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Song Han
- The First Hospital of Jilin University, Changchun, China
| | - Zhenxin Hou
- Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Chen Yang
- Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Yanbin Zhao
- Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| |
Collapse
|
38
|
YuYan, Yuan E. Regulatory effect of N6-methyladenosine on tumor angiogenesis. Front Immunol 2024; 15:1453774. [PMID: 39295872 PMCID: PMC11408240 DOI: 10.3389/fimmu.2024.1453774] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 08/19/2024] [Indexed: 09/21/2024] Open
Abstract
Previous studies have demonstrated that genetic alterations governing epigenetic processes frequently drive tumor development and that modifications in RNA may contribute to these alterations. In the 1970s, researchers discovered that N6-methyladenosine (m6A) is the most prevalent form of RNA modification in advanced eukaryotic messenger RNA (mRNA) and noncoding RNA (ncRNA). This modification is involved in nearly all stages of the RNA life cycle. M6A modification is regulated by enzymes known as m6A methyltransferases (writers) and demethylases (erasers). Numerous studies have indicated that m6A modification can impact cancer progression by regulating cancer-related biological functions. Tumor angiogenesis, an important and unregulated process, plays a pivotal role in tumor initiation, growth, and metastasis. The interaction between m6A and ncRNAs is widely recognized as a significant factor in proliferation and angiogenesis. Therefore, this article provides a comprehensive review of the regulatory mechanisms underlying m6A RNA modifications and ncRNAs in tumor angiogenesis, as well as the latest advancements in molecular targeted therapy. The aim of this study is to offer novel insights for clinical tumor therapy.
Collapse
Affiliation(s)
- YuYan
- Department of Laboratory Medicine, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Enwu Yuan
- Department of Laboratory Medicine, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
39
|
Kang K, Lin X, Chen P, Liu H, Liu F, Xiong W, Li G, Yi M, Li X, Wang H, Xiang B. T cell exhaustion in human cancers. Biochim Biophys Acta Rev Cancer 2024; 1879:189162. [PMID: 39089484 DOI: 10.1016/j.bbcan.2024.189162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 08/04/2024]
Abstract
T cell exhaustion refers to a progressive state in which T cells become functionally impaired due to sustained antigenic stimulation, which is characterized by increased expression of immune inhibitory receptors, but weakened effector functions, reduced self-renewal capacity, altered epigenetics, transcriptional programme and metabolism. T cell exhaustion is one of the major causes leading to immune escape of cancer, creating an environment that supports tumor development and metastatic spread. In addition, T cell exhaustion plays a pivotal role to the efficacy of current immunotherapies for cancer. This review aims to provide a comprehensive view of roles of T cell exhaustion in cancer development and progression. We summerized the regulatory mechanisms that involved in T cell exhaustion, including transcription factors, epigenetic and metabolic reprogramming events, and various microenvironmental factors such as cytokines, microorganisms, and tumor autocrine substances. The paper also discussed the challenges posed by T cell exhaustion to cancer immunotherapies, including immune checkpoint blockade (ICB) therapies and chimeric antigen receptor T cell (CAR-T) therapy, highlightsing the obstacles encountered in ICB therapies and CAR-T therapies due to T cell exhaustion. Finally, the article provides an overview of current therapeutic options aimed to reversing or alleviating T cell exhaustion in ICB and CAR-T therapies. These therapeutic approaches seek to overcome T cell exhaustion and enhance the effectiveness of immunotherapies in treating tumors.
Collapse
Affiliation(s)
- Kuan Kang
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, Hunan, China; The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha 410008, Hunan, China; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha 410078, Hunan, China
| | - Xin Lin
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, Hunan, China; The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha 410008, Hunan, China; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha 410078, Hunan, China
| | - Pan Chen
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, Hunan, China
| | - Huai Liu
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, Hunan, China; Department of Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China
| | - Feng Liu
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, Hunan, China; Department of Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China
| | - Wei Xiong
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, Hunan, China; The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha 410008, Hunan, China; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha 410078, Hunan, China
| | - Guiyuan Li
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, Hunan, China; The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha 410008, Hunan, China; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha 410078, Hunan, China
| | - Mei Yi
- Department of Dermatology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Xiayu Li
- Hunan Key Laboratory of Nonresolving Infammation and Cancer, The Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China.
| | - Hui Wang
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, Hunan, China; Department of Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China.
| | - Bo Xiang
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, Hunan, China; The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha 410008, Hunan, China; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha 410078, Hunan, China; FuRong Laboratory, Changsha 410078, Hunan, China.
| |
Collapse
|
40
|
Qu Y, Gao N, Zhang S, Gao L, He B, Wang C, Gong C, Shi Q, Li Z, Yang S, Xiao Y. Role of N6-methyladenosine RNA modification in cancer. MedComm (Beijing) 2024; 5:e715. [PMID: 39252821 PMCID: PMC11381670 DOI: 10.1002/mco2.715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 08/13/2024] [Accepted: 08/13/2024] [Indexed: 09/11/2024] Open
Abstract
N6-methyladenosine (m6A) is the most abundant modification of RNA in eukaryotic cells. Previous studies have shown that m6A is pivotal in diverse diseases especially cancer. m6A corelates with the initiation, progression, resistance, invasion, and metastasis of cancer. However, despite these insights, a comprehensive understanding of its specific roles and mechanisms within the complex landscape of cancer is still elusive. This review begins by outlining the key regulatory proteins of m6A modification and their posttranslational modifications (PTMs), as well as the role in chromatin accessibility and transcriptional activity within cancer cells. Additionally, it highlights that m6A modifications impact cancer progression by modulating programmed cell death mechanisms and affecting the tumor microenvironment through various cancer-associated immune cells. Furthermore, the review discusses how microorganisms can induce enduring epigenetic changes and oncogenic effect in microorganism-associated cancers by altering m6A modifications. Last, it delves into the role of m6A modification in cancer immunotherapy, encompassing RNA therapy, immune checkpoint blockade, cytokine therapy, adoptive cell transfer therapy, and direct targeting of m6A regulators. Overall, this review clarifies the multifaceted role of m6A modification in cancer and explores targeted therapies aimed at manipulating m6A modification, aiming to advance cancer research and improve patient outcomes.
Collapse
Affiliation(s)
- Yi Qu
- Department of Gastroenterology Xinqiao Hospital Army Medical University Chongqing China
| | - Nannan Gao
- Department of Gastroenterology Xinqiao Hospital Army Medical University Chongqing China
| | - Shengwei Zhang
- Department of Gastroenterology Xinqiao Hospital Army Medical University Chongqing China
| | - Limin Gao
- Department of Gastroenterology Xinqiao Hospital Army Medical University Chongqing China
| | - Bing He
- Department of Gastroenterology Xinqiao Hospital Army Medical University Chongqing China
| | - Chao Wang
- Department of Gastroenterology Xinqiao Hospital Army Medical University Chongqing China
| | - Chunli Gong
- Department of Gastroenterology Xinqiao Hospital Army Medical University Chongqing China
| | - Qiuyue Shi
- Department of Gastroenterology the First Affiliated Hospital of Guangxi Medical University Nanning Guangxi China
| | - Zhibin Li
- Department of Gastroenterology Xinqiao Hospital Army Medical University Chongqing China
| | - Shiming Yang
- Department of Gastroenterology Xinqiao Hospital Army Medical University Chongqing China
| | - Yufeng Xiao
- Department of Gastroenterology Xinqiao Hospital Army Medical University Chongqing China
| |
Collapse
|
41
|
Sun X, Wang H, Pu X, Wu Y, Yuan X, Wang X, Lu H. Manipulating the tumour immune microenvironment by N6-methyladenosine RNA modification. Cancer Gene Ther 2024; 31:1315-1322. [PMID: 38834772 DOI: 10.1038/s41417-024-00791-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 05/22/2024] [Accepted: 05/23/2024] [Indexed: 06/06/2024]
Abstract
N6-methyladenosine (m6A), a posttranscriptional regulatory mechanism, is the most common epigenetic modification in mammalian mRNA. M6A modifications play a crucial role in the developmental network of immune cells. The expression of m6A-related regulators often affects carcinogenesis and tumour suppression networks. In the tumour microenvironment, m6A-modified enzymes can affect the occurrence and progression of tumours by regulating the activation and invasion of tumour-associated immune cells. Immunotherapy, which utilises immune cells, has been demonstrated to be a powerful weapon in tumour treatment and is increasingly being used in the clinic. Here, we provide an updated and comprehensive overview of how m6A modifications affect invasive immune cells and their potential role in immune regulation. In addition, we summarise the regulation of epigenetic regulators associated with m6A modifications in tumour cells on the antitumour response of immune cells in the tumour immune microenvironment. These findings provide new insights into the role of m6A modifications in the immune response and tumour development, leading to the development of novel immunotherapies for cancer treatment.
Collapse
Affiliation(s)
- Xinyu Sun
- Department of Otorhinolaryngology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
- Department of Radiation Oncology, Cancer Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Huirong Wang
- Department of Otorhinolaryngology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Xi Pu
- Department of Radiation Oncology, Cancer Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yuting Wu
- Department of Radiation Oncology, Cancer Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Xiao Yuan
- Department of Radiation Oncology, Cancer Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Xu Wang
- Department of Radiation Oncology, Cancer Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Hanqiang Lu
- Department of Otorhinolaryngology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China.
| |
Collapse
|
42
|
Mu S, Zhao K, Zhong S, Wang Y. The Role of m6A Methylation in Tumor Immunity and Immune-Associated Disorder. Biomolecules 2024; 14:1042. [PMID: 39199429 PMCID: PMC11353047 DOI: 10.3390/biom14081042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 08/11/2024] [Accepted: 08/14/2024] [Indexed: 09/01/2024] Open
Abstract
N6-methyladenosine (m6A) represents the most prevalent and significant internal modification in mRNA, with its critical role in gene expression regulation and cell fate determination increasingly recognized in recent research. The immune system, essential for defense against infections and maintaining internal stability through interactions with other bodily systems, is significantly influenced by m6A modification. This modification acts as a key post-transcriptional regulator of immune responses, though its effects on different immune cells vary across diseases. This review delineates the impact of m6A modification across major system-related cancers-including those of the respiratory, digestive, endocrine, nervous, urinary reproductive, musculoskeletal system malignancies, as well as acute myeloid leukemia and autoimmune diseases. We explore the pathogenic roles of m6A RNA modifications within the tumor immune microenvironment and the broader immune system, highlighting how RNA modification regulators interact with immune pathways during disease progression. Furthermore, we discuss how the expression patterns of these regulators can influence disease susceptibility to immunotherapy, facilitating the development of diagnostic and prognostic models and pioneering new therapeutic approaches. Overall, this review emphasizes the challenges and prospective directions of m6A-related immune regulation in various systemic diseases throughout the body.
Collapse
Affiliation(s)
- Siyu Mu
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang 110000, China; (S.M.); (S.Z.)
| | - Kaiyue Zhao
- Department of Hepatology, Beijing Tsinghua Changgeng Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China;
| | - Shanshan Zhong
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang 110000, China; (S.M.); (S.Z.)
| | - Yanli Wang
- Department of Infectious Diseases, The First Affiliated Hospital of China Medical University, Shenyang 110000, China
| |
Collapse
|
43
|
Li F, Zeng C, Liu J, Wang L, Yuan X, Yuan L, Xia X, Huang W. The YTH domain-containing protein family: Emerging players in immunomodulation and tumour immunotherapy targets. Clin Transl Med 2024; 14:e1784. [PMID: 39135292 PMCID: PMC11319238 DOI: 10.1002/ctm2.1784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 07/12/2024] [Accepted: 07/16/2024] [Indexed: 08/16/2024] Open
Abstract
BACKGROUND The modification of N6-methyladenosine (m6A) plays a pivotal role in tumor by altering both innate and adaptive immune systems through various pathways, including the regulation of messenger RNA. The YTH domain protein family, acting as "readers" of m6A modifications, affects RNA splicing, stability, and immunogenicity, thereby playing essential roles in immune regulation and antitumor immunity. Despite their significance, the impact of the YTH domain protein family on tumor initiation and progression, as well as their involvement in tumor immune regulation and therapy, remains underexplored and lacks comprehensive review. CONCLUSION This review introduces the molecular characteristics of the YTH domain protein family and their physiological and pathological roles in biological behavior, emphasizing their mechanisms in regulating immune responses and antitumor immunity. Additionally, the review discusses the roles of the YTH domain protein family in immune-related diseases and tumor resistance, highlighting that abnormal expression or dysfunction of YTH proteins is closely linked to tumor resistance. KEY POINTS This review provides an in-depth understanding of the YTH domain protein family in immune regulation and antitumor immunity, suggesting new strategies and directions for immunotherapy of related diseases. These insights not only deepen our comprehension of m6A modifications and YTH protein functions but also pave the way for future research and clinical applications.
Collapse
Affiliation(s)
- Fenghe Li
- Department of Gynaecology and ObstetricsSecond Xiangya HospitalCentral South UniversityChangshaChina
| | - Chong Zeng
- Department of Respiratory and Critical Care MedicineThe Seventh Affiliated Hospital, Hengyang Medical School, University of South ChinaChangshaHunanChina
| | - Jie Liu
- Department of PathologyThe Affiliated Changsha Central Hospital, Hengyang Medical School, University of South ChinaChangshaHunanChina
| | - Lei Wang
- NHC Key Laboratory of Carcinogenesis and the Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of EducationCancer Research Institute, School of Basic Medical Science, Central South UniversityChangshaHunanChina
| | - Xiaorui Yuan
- Department of Gynaecology and ObstetricsSecond Xiangya HospitalCentral South UniversityChangshaChina
| | - Li Yuan
- Department of Nuclear MedicineThe Third Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Xiaomeng Xia
- Department of Gynaecology and ObstetricsSecond Xiangya HospitalCentral South UniversityChangshaChina
| | - Wei Huang
- Department of OncologyXiangya HospitalCentral South UniversityChangshaChina
- National Clinical Research Center of Geriatric DisordersXiangya HospitalCentral South UniversityChangshaChina
- Research Center of Carcinogenesis and Targeted TherapyXiangya HospitalCentral South UniversityChangshaChina
| |
Collapse
|
44
|
Sun HJ, Zheng ZF, Zhang LJ, Fang L, Fu H, Chen SY, Feng RX, Liu XY, Tang QN, Liu XW. Increased infiltration of M2-polarized tumour-associated macrophages is highly associated with advanced disease stage and high expression of PD-L1 in buccal mucosa carcinoma. Discov Oncol 2024; 15:314. [PMID: 39073672 PMCID: PMC11286931 DOI: 10.1007/s12672-024-01190-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 07/23/2024] [Indexed: 07/30/2024] Open
Abstract
OBJECTIVE To assess the infiltration characteristics of tumour-associated macrophages (TAMs) in buccal mucosa carcinoma (BMC) and the correlation of these features with clinicopathological factors. MATERIALS AND METHODS Immunohistochemistry was used to detect the expression of TAM-related markers (CD68, CD163, CD206), CD8+ T cell markers, PD-L1, and epidermal growth factor receptor (EGFR) in 46 patients with mucosal cancer after radical surgery. In addition, the correlation between TAM infiltration and clinical characteristics, PD-L1 expression, and EGFR expression was analysed. RESULTS A high infiltration level of M2-polarized (CD206+) TAMs and M2-polarized (CD163+) TAMs was more common in stage T3-T4, N+, III-IV patients than in other patient groups (P < 0.05). The infiltration degree of M2-polarized (CD68+) TAMs was positively correlated with the PD-L1 TPS (P = 0.0331). The infiltration level of M2-polarized (CD206+) TAMs was higher in the EGFR high expression group than in the EGFR low expression group (P = 0.040). CONCLUSION High infiltration of M2-polarized TAMs is highly associated with advanced disease stage and higher expression of PD-L1 and EGFR in BMCs, suggesting that M2-polarized TAMs infiltration can serve as a potential therapeutic target.
Collapse
Affiliation(s)
- Hao-Jia Sun
- Department of Oncology, Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Zhui-Feng Zheng
- Department of Oncology, Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
- Department of Breast Medical Oncology, Fujian Cancer Hospital and the Fujian Medical University Cancer Hospital, Fuzhou, 350014, Fujian, China
| | - Li-Jun Zhang
- Department of Oncology, Huaihua First People's Hospital, Huaihua, 418000, Hunan, China
| | - Le Fang
- Department of Oncology, Loudi Central Hospital, Loudi, 417099, Hunan, China
| | - Hua Fu
- Department of Pathology, Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Shao-Yang Chen
- Department of Oncology, Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Rong-Xiu Feng
- Department of Radiation Oncology, Xiangtan Central Hospital, Xiangtan, 411199, Hunan, China
| | - Xiao-Yang Liu
- Department of Oncology, Changde First People's Hospital, Changde, 415003, Hunan, China
| | - Qing-Nan Tang
- Department of Oncology, Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Xue-Wen Liu
- Department of Oncology, Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China.
| |
Collapse
|
45
|
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: 8] [Impact Index Per Article: 8.0] [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
|
46
|
Liu W, Kuang T, Liu L, Deng W. The role of innate immune cells in the colorectal cancer tumor microenvironment and advances in anti-tumor therapy research. Front Immunol 2024; 15:1407449. [PMID: 39100676 PMCID: PMC11294098 DOI: 10.3389/fimmu.2024.1407449] [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: 03/26/2024] [Accepted: 06/25/2024] [Indexed: 08/06/2024] Open
Abstract
Innate immune cells in the colorectal cancer microenvironment mainly include macrophages, neutrophils, natural killer cells, dendritic cells and bone marrow-derived suppressor cells. They play a pivotal role in tumor initiation and progression through the secretion of diverse cytokines, chemokines, and other factors that govern these processes. Colorectal cancer is a common malignancy of the gastrointestinal tract, and understanding the role of innate immune cells in the microenvironment of CRC may help to improve therapeutic approaches to CRC and increase the good prognosis. In this review, we comprehensively explore the pivotal role of innate immune cells in the initiation and progression of colorectal cancer (CRC), alongside an extensive evaluation of the current landscape of innate immune cell-based immunotherapies, thereby offering valuable insights for future research strategies and clinical trials.
Collapse
Affiliation(s)
| | | | | | - Wenhong Deng
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
47
|
Xu R, Liu X, Zhang Y, Wu G, Huang L, Li R, Xu X. Antibody-Decorated Nanoplatform to Reprogram Macrophage and Block Immune Checkpoint LSECtin for Effective Cancer Immunotherapy. NANO LETTERS 2024; 24:8723-8731. [PMID: 38968148 DOI: 10.1021/acs.nanolett.4c02139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/07/2024]
Abstract
Repolarizing tumor-associated macrophages (TAMs) into tumor-inhibiting M1 macrophages has been considered a promising strategy for enhanced cancer immunotherapy. However, several immunosuppressive ligands (e.g., LSECtin) can still be highly expressed on M1 macrophages, inducing unsatisfactory therapeutic outcomes. We herein developed an antibody-decorated nanoplatform composed of PEGylated iron oxide nanoparticles (IONPs) and LSECtin antibody conjugated onto the surface of IONPs via the hydrazone bond for enhanced cancer immunotherapy. After intravenous administration, the tumor microenvironment (TME) pH could trigger the hydrazone bond breakage and induce the disassociation of the nanoplatform into free LSECtin antibodies and IONPs. Consequently, the IONPs could repolarize TAMs into M1 macrophages to remodel immunosuppressive TME and provide an additional anticancer effect via secreting tumoricidal factors (e.g., interlukin-12). Meanwhile, the LSECtin antibody could further block the activity of LSECtin expressed on M1 macrophages and relieve its immunosuppressive effect on CD8+ T cells, ultimately leading to significant inhibition of tumor growth.
Collapse
Affiliation(s)
- Rui Xu
- Department of Pharmacy and Pharmacology and the Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, P. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P. R. China
- Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P. R. China
| | - Xiangya Liu
- Department of Pharmacy and Pharmacology and the Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, P. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P. R. China
- Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P. R. China
| | - Yuxuan Zhang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P. R. China
- Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P. R. China
| | - Guo Wu
- Department of Pharmacy and Pharmacology and the Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, P. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P. R. China
- Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P. R. China
| | - Linzhuo Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P. R. China
- Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P. R. China
| | - Rong Li
- Department of Pharmacy and Pharmacology and the Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, P. R. China
| | - Xiaoding Xu
- Department of Pharmacy and Pharmacology and the Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, P. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P. R. China
- Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P. R. China
| |
Collapse
|
48
|
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
|
49
|
Gu J, Xu X, Li X, Yue L, Zhu X, Chen Q, Gao J, Takashi M, Zhao W, Zhao B, Zhang Y, Lin M, Zhou J, Liang Y, Dai S, Pan Y, Shao Q, Li Y, Wang Y, Xu Z, Qian Q, Huang T, Qian X, Lu L. Tumor-resident microbiota contributes to colorectal cancer liver metastasis by lactylation and immune modulation. Oncogene 2024; 43:2389-2404. [PMID: 38890429 PMCID: PMC11281901 DOI: 10.1038/s41388-024-03080-7] [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: 02/28/2024] [Revised: 05/31/2024] [Accepted: 06/05/2024] [Indexed: 06/20/2024]
Abstract
The role of tumor-resident microbiota in modulating tumor immunity remains unclear. Here, we discovered an abundance of intra-tumoral bacteria, such us E.coli, residing and resulting in Colorectal cancer liver metastasis (CRLM). E.coli enhanced lactate production, which mediated M2 macrophage polarization by suppressing nuclear factor-κB -gene binding (NF-κB) signaling through retinoic acid-inducible gene 1 (RIG-I) lactylation. Lactylation of RIG-I suppressed recruitment of NF-κB to the Nlrp3 promoter in macrophages, thereby reducing its transcription. This loss of Nlrp3 affected the immunosuppressive activities of regulatory T cells (Tregs) and the antitumor activities of and CD8+ T cells. Small-molecule compound screening identified a RIG-I lactylation inhibitor that suppressed M2 polarization and sensitized CRLM to 5-fluorouracil (5-FU). Our findings suggest that tumor-resident microbiota may be a potential target for preventing and treating CRLM.
Collapse
Affiliation(s)
- Jian Gu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University and Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China.
- Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.
| | - Xiaozhang Xu
- Department of General Surgery, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, China
| | - Xiangyu Li
- Department of General Surgery, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, China
| | - Lei Yue
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
| | - Xiaowen Zhu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University and Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Qiuyang Chen
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University and Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Ji Gao
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University and Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | | | - Wenhu Zhao
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University and Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Bo Zhao
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, China
| | - Yue Zhang
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, China
| | - Minjie Lin
- The Clinical Skills Training Center, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Jinren Zhou
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University and Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Yuan Liang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University and Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
- School of Biological Science & Medical Engineering, Southeast University, Nanjing, China
| | - Shipeng Dai
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University and Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Yufeng Pan
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University and Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
- School of Medicine, Southeast University, Nanjing, China
| | - Qing Shao
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University and Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Yu Li
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University and Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Yiming Wang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University and Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Zibo Xu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University and Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Qufei Qian
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University and Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Tianning Huang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University and Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Xiaofeng Qian
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University and Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China.
- Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.
| | - Ling Lu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University and Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China.
- Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.
- Department of General Surgery, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
50
|
Yang X, Tang H, Sun X, Gui Q. M6A modification and T cells in adipose tissue inflammation. Cell Biochem Funct 2024; 42:e4089. [PMID: 38978329 DOI: 10.1002/cbf.4089] [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: 03/04/2024] [Revised: 05/11/2024] [Accepted: 06/28/2024] [Indexed: 07/10/2024]
Abstract
Adipose tissue in the obese state can lead to low-grade chronic inflammation while inducing or exacerbating obesity-related metabolic diseases and impairing overall health.T cells, which are essential immune cells similar to macrophages, are widely distributed in adipose tissue and perform their immunomodulatory function; they also cross-talk with other cells in the vascular stromal fraction. Based on a large number of studies, it has been found that N6 methyl adenine (m6A) is one of the most representative of epigenetic modifications, which affects the crosstalk between T cells, as well as other immune cells, in several ways and plays an important role in the development of adipose tissue inflammation and related metabolic diseases. In this review, we first provide an overview of the widespread presence of T cells in adipose tissue and summarize the key role of T cells in adipose tissue inflammation. Next, we explored the effects of m6A modifications on T cells in adipose tissue from the perspective of adipose tissue inflammation. Finally, we discuss the impact of m6a-regulated crosstalk between T cells and immune cells on the prospects for improving adipose tissue inflammation research, providing additional new ideas for the treatment of obesity.
Collapse
Affiliation(s)
- Xiaoting Yang
- Institute of Translational Medicine, Department of Basic Medicine, Hengyang Medical School, University of South China, Hengyang, China
| | - Haojun Tang
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, China
| | - Xuan Sun
- Institute of Translational Medicine, Department of Basic Medicine, Hengyang Medical School, University of South China, Hengyang, China
| | - Qingjun Gui
- Institute of Translational Medicine, Department of Basic Medicine, Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|