1
|
Lan J, Cai D, Gou S, Bai Y, Lei H, Li Y, Chen Y, Zhao Y, Shen J, Wu X, Li M, Chen M, Li X, Sun Y, Gu L, Li W, Wang F, Cho CH, Zhang Y, Zheng X, Xiao Z, Du F. The dynamic role of ferroptosis in cancer immunoediting: Implications for immunotherapy. Pharmacol Res 2025; 214:107674. [PMID: 40020885 DOI: 10.1016/j.phrs.2025.107674] [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: 12/19/2024] [Revised: 02/14/2025] [Accepted: 02/23/2025] [Indexed: 03/03/2025]
Abstract
Currently, cancer immunotherapy strategies are primarily formulated based on the patient's present condition, representing a "static" treatment approach. However, cancer progression is inherently "dynamic," as the immune environment is not fixed but undergoes continuous changes. This dynamism is characterized by the ongoing interactions between tumor cells and immune cells, which ultimately lead to alterations in the tumor immune microenvironment. This process can be effectively elucidated by the concept of cancer immunoediting, which divides tumor development into three phases: "elimination," "equilibrium," and "escape." Consequently, adjusting immunotherapy regimens based on these distinct phases may enhance patient survival and improve prognosis. Targeting ferroptosis is an emerging area in cancer immunotherapy, and our findings reveal that the antioxidant systems associated with ferroptosis possess dual roles, functioning differently across the three phases of cancer immunoediting. Therefore, this review delve into the dual role of the ferroptosis antioxidant system in tumor development and progression. It also propose immunotherapy strategies targeting ferroptosis at different stages, ultimately aiming to illuminate the significant implications of targeting ferroptosis at various phases for cancer immunotherapy.
Collapse
Affiliation(s)
- Jiarui Lan
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646600, China
| | - Dan Cai
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646600, China
| | - Shuang Gou
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China
| | - Yulin Bai
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China
| | - Huaqing Lei
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646600, China
| | - Yan Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China
| | - Yu Chen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646600, China
| | - Yueshui Zhao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646600, China
| | - Jing Shen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646600, China
| | - Xu Wu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646600, China
| | - Mingxing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646600, China
| | - Meijuan Chen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China
| | - Xiaobing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China
| | - Yuhong Sun
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China
| | - Li Gu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China
| | - Wanping Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China
| | - Fang Wang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China
| | - Chi Hin Cho
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Yan Zhang
- Department of Oncology, Luzhou People's Hospital, Luzhou, Sichuan 646000, China
| | - Xin Zheng
- Department of Oncology, Luzhou People's Hospital, Luzhou, Sichuan 646000, China.
| | - Zhangang Xiao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646600, China.
| | - Fukuan Du
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646600, China.
| |
Collapse
|
2
|
Wang X, Chen L, Zhang W, Sun W, Huang J. Colorectal Cancer-Derived Exosomes Impair CD4 + T Cell Function and Accelerate Cancer Progression via Macrophage Activation. Cancer Biother Radiopharm 2025; 40:185-195. [PMID: 39263734 DOI: 10.1089/cbr.2024.0032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2024] Open
Abstract
Background: Exosomal programmed death ligand 1 (PD-L1), an exosomal membrane protein found in many tumor types, is consider to aid in regulation of the immune microenvironment. However, the functions and the mechanisms underlying the exosome-mediated regulation of the immune microenvironment in colorectal cancer (CRC) remain unknown. Methods: Western blotting was used to investigate the levels of exosomal PD-L1 in the peripheral blood of patients with CRC and healthy controls. A CRC mouse model was constructed by administering 10 mg/kg azoxymethane (AOM) and dextrane sodium sulfate (DSS) intraperitoneally. The mice were then administered the control or CRC-derived exosomes to examine the regulatory effect of the exosomes on macrophage infiltration and CRC development. In vitro studies, using a coculture system, and flow cytometry analysis were conducted to examine the relationship between the regulatory effect of CRC-derived exosomes on CD4+ T cells and tumor-associated macrophages. RNA-seq and reverse transcription-quantitative polymerase chain reaction assays were used to investigate the mechanisms underlying the regulatory effect of the CRC-derived exosomes on macrophage proliferation and the regulation of the immune microenvironment during CRC development. Results: In patients with CRC, higher levels of exosomal PD-L1 were associated with a more severe form of disease. The treatment of mice with AOM/DSS-induced CRC with CRC-derived exosomes resulted in high levels of macrophage proliferation, increased PD-L1 levels in macrophages, and accelerated CRC progression. Importantly, analysis of an in vitro coculture system and flow cytometry analysis showed that the CRC-derived exosomes transported PD-L1 into macrophages and impaired CD4+ T cell function. Preliminary data suggest that the NF-κb signaling pathway regulates the function of CRC-derived exosomal PD-L1-dependent macrophages. Conclusion: CRC-derived exosomes induce the proliferation of macrophages and increase their PD-L1 levels. They also impair CD4+ T cell function and promote CRC progression. Our findings reveal a novel exosomal PD-L1-mediated crosstalk between the CRC cells and immune cells in the CRC microenvironment.
Collapse
Affiliation(s)
- Xiaolong Wang
- Department of Gastrointestinal Surgery, Huadu District People's Hospital of Guangzhou, Guangzhou, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Liang Chen
- Department of Gastrointestinal Surgery, Huadu District People's Hospital of Guangzhou, Guangzhou, China
| | - Wenwei Zhang
- Department of Gastrointestinal Surgery, Huadu District People's Hospital of Guangzhou, Guangzhou, China
| | - Wei Sun
- Department of Gastrointestinal Surgery, Huadu District People's Hospital of Guangzhou, Guangzhou, China
| | - Jianpeng Huang
- Department of Gastrointestinal Surgery, The Third People's Hospital of Shenzhen, Shenzhen, China
| |
Collapse
|
3
|
Almawash S. Revolutionary Cancer Therapy for Personalization and Improved Efficacy: Strategies to Overcome Resistance to Immune Checkpoint Inhibitor Therapy. Cancers (Basel) 2025; 17:880. [PMID: 40075727 PMCID: PMC11899125 DOI: 10.3390/cancers17050880] [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: 01/06/2025] [Revised: 02/09/2025] [Accepted: 02/12/2025] [Indexed: 03/14/2025] Open
Abstract
Cancer remains a significant public health issue worldwide, standing as a primary contributor to global mortality, accounting for approximately 10 million fatalities in 2020 [...].
Collapse
Affiliation(s)
- Saud Almawash
- Department of Pharmaceutics, College of Pharmacy, Shaqra University, Shaqra 11961, Saudi Arabia
| |
Collapse
|
4
|
Zhu L, Qiu X, Liang S, Huang S, Ning Q, Chen X, Chen N, Qin L, Huang J, Liu S. Identification of a novel signature based on RNA methylation-associated anoikis-related genes for predicting prognosis and characterizing immune landscape in colorectal cancer. Discov Oncol 2025; 16:239. [PMID: 40000539 PMCID: PMC11861771 DOI: 10.1007/s12672-025-01964-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: 09/09/2024] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND RNA methylation is a potential target for cancer therapy, while anoikis, a form of programmed cell death, is linked to cancer metastasis. However, the prognostic and immune significance of RNA methylation- and anoikis-related genes in colorectal cancer (CRC) remains unknown. METHODS Transcriptomic and clinicopathological data for CRC were obtained from TCGA and the GEO databases. A novel signature was constructed based on RNA methylation- and anoikis-related genes using univariate and multivariate Cox regression as well as LASSO Cox regression methods. CRC patients were stratified into low- and high-risk groups based on this signature. Differences in prognosis, immune infiltration, and drug sensitivity between two groups were analyzed. Finally, immunohistochemistry, western blot, and RT-qPCR were employed to validate the expression of the key gene SERPINE1 in CRC tissues and cells, as well as the effect of FTO on its expression. RESULTS We identified 79 differentially expressed RNA methylation-associated anoikis-related genes (RMRARGs) in both cancerous and normal tissues. A signature composed of 9 key genes (BID, FASN, PLK1, CDKN3, MYC, EPHA2, SERPINE1, CD36, PDK4) was established. Kaplan-Meier analysis revealed a poorer prognosis in the high-risk group. Compared to the other three published models, this signature demonstrated superior predictive performance based on the ROC curve analysis. Functional analyses highlighted differences in drug sensitivities and signaling pathways between risk groups. Furthermore, immune analysis results showed that risk score was associated with some immune cells and immune checkpoints. Immunohistochemistry showed high SERPINE1 expression in CRC tissues, with FTO expression positively correlated with SERPINE1. Furthermore, RT-qPCR and western blot indicated FTO knockdown markedly downregulated SERPINE1 levels. CONCLUSION Our findings underscore the prognostic value of this signature in CRC patients and its utility in assessing immune status. Additionally, the m6A demethylase FTO regulates the expression of the anoikis-related gene SERPINE1.
Collapse
Affiliation(s)
- Liye Zhu
- Department of Gastroenterology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, Guangxi, China
| | - Xinze Qiu
- Department of Gastroenterology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, Guangxi, China
| | - Shengmei Liang
- Department of Gastroenterology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, Guangxi, China
| | - Shanpei Huang
- Department of Gastroenterology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, Guangxi, China
| | - Qiting Ning
- Department of Gastroenterology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, Guangxi, China
| | - Xingmei Chen
- Department of Gastroenterology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, Guangxi, China
| | - Ni Chen
- Department of Gastroenterology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, Guangxi, China
| | - Longjie Qin
- Department of Gastroenterology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, Guangxi, China
| | - Jiean Huang
- Department of Gastroenterology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, Guangxi, China.
| | - Shiquan Liu
- Department of Gastroenterology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, Guangxi, China.
| |
Collapse
|
5
|
Ge Y, Zhou Q, Pan F, Wang R. Utilizing Nanoparticles to Overcome Anti-PD-1/PD-L1 Immunotherapy Resistance in Non-Small Cell Lung cancer: A Potential Strategy. Int J Nanomedicine 2025; 20:2371-2394. [PMID: 40027868 PMCID: PMC11871910 DOI: 10.2147/ijn.s505539] [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: 11/21/2024] [Accepted: 01/25/2025] [Indexed: 03/05/2025] Open
Abstract
Lung cancer is the leading cause of cancer-related mortality globally, with non-small cell lung cancer (NSCLC) constituting 85% of cases. Immune checkpoint inhibitors (ICIs) represented by anti-programmed cell death protein 1 (PD-1)/ programmed cell death ligand 1 (PD-L1) have emerged as a promising frontier in cancer treatment, effectively extending the survival of patients with NSCLC. However, the efficacy of ICIs exhibits significant variability across diverse patient populations, with a substantial proportion showing poor responsiveness and acquired resistance in those initially responsive to ICIs treatments. With the advancement of nanotechnology, nanoparticles offer unique advantages in tumor immunotherapy, including high permeability and prolonged retention(EPR) effects, enhanced drug delivery and stability, and modulation of the inflammatory tumor microenvironment(TME). This review summarizes the mechanisms of resistance to ICIs in NSCLC, focusing on tumor antigens loss and defective antigen processing and presentation, failure T cell priming, impaired T cell migration and infiltration, immunosuppressive TME, and genetic mutations. Furthermore, we discuss how nanoparticles, through their intrinsic properties such as the EPR effect, active targeting effect, shielding effect, self-regulatory effect, and synergistic effect, can potentiate the efficacy of ICIs and reverse resistance. In conclusion, nanoparticles serve as a robust platform for ICIs-based NSCLC therapy, aiding in overcoming resistance challenges.
Collapse
Affiliation(s)
- Yuli Ge
- Department of Medical Oncology, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, People’s Republic of China
| | - Qiong Zhou
- Department of Medical Oncology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, 210093, People’s Republic of China
| | - Fan Pan
- Department of Medical Oncology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, 210093, People’s Republic of China
| | - Rui Wang
- Department of Medical Oncology, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, People’s Republic of China
| |
Collapse
|
6
|
Qiu H, Liu F, Qiu M, Yang J, Peng X. Monotropein attenuates renal cell carcinoma cell progression and M2 macrophage polarization by weakening NF-κB. Int Urol Nephrol 2025:10.1007/s11255-024-04358-1. [PMID: 39776402 DOI: 10.1007/s11255-024-04358-1] [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: 11/07/2024] [Accepted: 12/24/2024] [Indexed: 01/11/2025]
Abstract
PURPOSE The study aimed to investigate the effect and mechanism of monotropein on renal cell carcinoma (RCC). METHODS After monotropein and NF-κB receptor activator (RANKL) treatment, cell proliferation, invasion, and apoptosis were evaluated using CCK-8, Transwell, and flow cytometry. Primary macrophages co-cultured with monotropein-treated RCC cells were analyzed to evaluate macrophage polarization using qRT-PCR, western blot, and ELISA assays by detecting the expression of M2 markers (CD206, CD168) and cytokines (IL-10, TGF-β). Additionally, the therapeutic efficacy of monotropein was examined using an RCC mouse xenograft model. RESULTS Monotropein could inhibit the proliferation, invasion, and M2 macrophage polarization and accelerate the apoptosis of RCC cells. Mechanistically, monotropein suppressed NF-κB pathway activation in RCC cells and reduced the expression of NF-κB downstream targets, including Bcl-2, c-Myc, and MMP9. RANKL could eliminate the effect of monotropein on RCC progression. In primary macrophages co-cultured with monotropein-treated RCC cells, monotropein downregulated M2 polarization markers and cytokines, further supporting its role in modulating the tumor microenvironment. In mouse models, monotropein reduced RCC tumor growth, induced apoptosis, and blocked NF-κB pathway. CONCLUSIONS Monotropein prevents RCC malignant progression and reduces M2 macrophage polarization by suppressing the NF-κB pathway, suggesting that monotropein may serve as a potential therapeutic agent for RCC by targeting both tumor cells and the tumor microenvironment.
Collapse
Affiliation(s)
- Heping Qiu
- Department of Urology, The Second Affiliated Hospital of Nanchang University, No.1, Minde Road, East Lake District, Nanchang, 330006, Jiangxi, China
| | - Fei Liu
- Department of Urology, The Second Affiliated Hospital of Nanchang University, No.1, Minde Road, East Lake District, Nanchang, 330006, Jiangxi, China
| | - Mei Qiu
- Department of Urology, Shanghai Changzheng Hospital, Shanghai, 341099, China
| | - Juliang Yang
- Department of Urology, The Affiliated Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou, 341099, China
| | - Xiang Peng
- Department of Urology, The Second Affiliated Hospital of Nanchang University, No.1, Minde Road, East Lake District, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
7
|
Feng C, Chen R, Gao X, Fang W, Wu S, Chen L, Zheng X, Ji X, Yuan M, Fu Y, Ying H, Shen T, Zhu D, Jiang J. Cordycepin enhances the Anticancer efficacy of PD-L1 blockade by modulating the tumor microenvironment of colon cancer. Eur J Pharmacol 2024; 985:177089. [PMID: 39489279 DOI: 10.1016/j.ejphar.2024.177089] [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: 10/30/2024] [Accepted: 10/31/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND PD-L1 blockade has been found to be effective in treating multiple malignancies. Combined therapy is proposed to provide better therapeutic effects. Cordycepin, a prominent bioactive compound found in cordyceps, can inhibit the development of various cancers. PURPOSE This study aimed to determine the efficacy of combined anti-PD-L1 antibody and cordycepin in tumor treatment. METHODS A single-cell RNA sequencing was used to analyze the mechanism of combined treatment. RESULTS Combination therapy of anti-PD-L1 and cordycepin significantly inhibited tumor growth by regulating the T cell ratio and improving the function of CD8+T cells. Furthermore, cordycepin promoted the reprogramming of type-II macrophages into type-I macrophages, a process confirmed through flow cytometry analysis of the underlying mechanism. CONCLUSION Our findings demonstrate that the combination of anti-PD-L1 and cordycepin effectively suppressed tumor growth by regulating the proportion of T cells and reprograming type-II macrophages.
Collapse
Affiliation(s)
- Chen Feng
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, China; Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, China.
| | - Rongzhang Chen
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, China; Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, China.
| | - Xinran Gao
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, China; Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, China.
| | - Weiwei Fang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, China; Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, China.
| | - Shaoxian Wu
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, China; Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, China.
| | - Lujun Chen
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, China; Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, China.
| | - Xiao Zheng
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, China; Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, China.
| | - Xinyue Ji
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, China; Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, China.
| | - Maoling Yuan
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, China; Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, China.
| | - Yuanyuan Fu
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, China; Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, China; Department of Gynecology, Changzhou Traditional Chinese Medicine Hospital, Changzhou, Jiangsu, 213003, China.
| | - Hanjie Ying
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, Jiangsu, 211816, China; Soochow University, Suzhou, Jiangsu, 215031, China.
| | - Tao Shen
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, Jiangsu, 211816, China.
| | - Dawei Zhu
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, China; Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, China.
| | - Jingting Jiang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, China; Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, China.
| |
Collapse
|
8
|
Wang X, Zhang Y. Multi-omics joint screening of biomarkers related to M2 macrophages in gastric cancer. Discov Oncol 2024; 15:738. [PMID: 39623254 PMCID: PMC11612128 DOI: 10.1007/s12672-024-01623-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 11/25/2024] [Indexed: 12/06/2024] Open
Abstract
BACKGROUND Due to high mortality rate and limited treatments in gastric cancer (GC), call for deeper exploration of M2 macrophages as biomarkers is needed. METHODS The data for this study were obtained from the Gene Expression Omnibus (GEO) and Genomic Data Commons (GDC). The Seurat package was utilized for single-cell RNA sequencing (scRNA-seq) analysis. FindAllMarkers was used to identify genes highly expressed among different cell subsets. DESeq2 package was leveraged to screen differentially expressed genes (DEGs), while limma package was utilized for identifying differentially expressed proteins (DEPs). Enrichment analyses of the genes were conducted using KOBAS-i database. MultipleROC was applied to evaluate the diagnostic potential of biomarkers, and rms package was utilized to construct diagnostic models. hTFtarget database was utilized to predict potential transcription factors (TFs). Finally, cell-based assays were performed to validate the expression and potential biological functions of the screened key markers. RESULTS This study found that M2 macrophages were enriched in protein, endoplasmic reticulum, and virus-related pathways. A total of 4146 DEGs and 1946 DEPs were obtained through screening, with 254 common DEGs/DEPs. The results of gene function enrichment analysis suggested that it may affect the occurrence and development of GC through DNA replication and cell cycle. This study identified three biomarkers, HSPH1, HSPD1, and IFI30, and constructed a diagnostic model based on these three genes. The AUC value greater than 0.8 proved the reliability of the model. Through screening TFs, SPI1 and KLF5 were found to be the common TFs for the three biomarkers. The expression of the three genes IFI30, HSPD1 and HSPH1 was up-regulated in GC cells, and IFI30 may play a facilitating role in the migration and invasion of GC cells. CONCLUSION This study identified three biomarkers and constructed a diagnostic model, providing a new perspective for the research and treatment of GC.
Collapse
Affiliation(s)
- Xilong Wang
- Tumor Hematology Department, Liaoyang Central Hospital, Liaoyang, 111000, China
| | - Ying Zhang
- General Surgery Department, Liaoyang Central Hospital, Liaoyang, 111000, China.
| |
Collapse
|
9
|
Chen YH, Lee YM, Ou DL, Hsu CL, Hsu C, Chen CN, Ko JY, Tan CT. Regorafenib enhances M1/M2 macrophage polarization by inhibiting the secretion of plasminogen activator inhibitor-1 in head and neck squamous cell carcinoma. Life Sci 2024; 358:123147. [PMID: 39419267 DOI: 10.1016/j.lfs.2024.123147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/28/2024] [Accepted: 10/11/2024] [Indexed: 10/19/2024]
Abstract
AIMS Regorafenib, an oral multikinase inhibitor, is approved for the treatment of various metastatic/advanced cancers. Although clinical trials have reported the efficacy of regorafenib in multiple cancer types, its immunomodulatory activity in head and neck squamous cell carcinoma (HNSCC) remains unclear. MAIN METHODS This study investigated the effects of regorafenib on tumorigenesis by using two mouse models of HNSCC. The distribution of immune cells in tumor tissues was assessed through flow cytometry, RNA sequencing, and multiplex immunofluorescence staining. KEY FINDINGS Regorafenib exhibited significant antitumor activity in our HNSCC mouse models. Tumor-infiltrating lymphocyte isolation and RNA sequencing revealed that regorafenib can activate immune functions. Moreover, regorafenib-treated tumor-conditioned medium regulated macrophage proliferation ex vivo. Our data suggests that regorafenib modulates immune function by regulating both tumor and immune cells. Specifically, regorafenib induced the polarization of macrophages toward the proinflammatory M1 phenotype by suppressing the production of plasminogen activator inhibitor 1 (PAI-1), a macrophage regulator. In addition, regorafenib suppressed the secretion of PAI-1 from ex vivo human HNSCC organoids. SIGNIFICANCE Regorafenib enhances M1/M2 macrophage polarization and suppresses PAI-1 secretion from cancer cells, leading to a shift from M2 to M1 macrophages in the HNSCC tumor microenvironment.
Collapse
Affiliation(s)
- Yu-Hsin Chen
- Department of Otolaryngology, National Taiwan University Hospital, Taipei 10051, Taiwan; Stem Cell Core Laboratory, Center of Genomic Medicine, National Taiwan University, Taipei 10051, Taiwan; Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei 10051, Taiwan
| | - Yi-Mei Lee
- Department of Otolaryngology, National Taiwan University Hospital, Taipei 10051, Taiwan; Stem Cell Core Laboratory, Center of Genomic Medicine, National Taiwan University, Taipei 10051, Taiwan
| | - Da-Liang Ou
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei 10051, Taiwan; YongLin Institute of Health, National Taiwan University, Taipei 10051, Taiwan
| | - Chia-Lang Hsu
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei 10051, Taiwan; Department of Medical Research, National Taiwan University Hospital, Taipei 10051, Taiwan
| | - Chiun Hsu
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei 10051, Taiwan; Department of Oncology, National Taiwan University Cancer Center, Taipei 10672, Taiwan; Department of Oncology, National Taiwan University Hospital, Taipei 10051, Taiwan
| | - Chun-Nan Chen
- Department of Otolaryngology, National Taiwan University Hospital, Taipei 10051, Taiwan
| | - Jenq-Yuh Ko
- Department of Otolaryngology, National Taiwan University Hospital, Taipei 10051, Taiwan
| | - Ching-Ting Tan
- Department of Otolaryngology, National Taiwan University Hospital, Taipei 10051, Taiwan; Stem Cell Core Laboratory, Center of Genomic Medicine, National Taiwan University, Taipei 10051, Taiwan; Department of Otolaryngology, National Taiwan University College of Medicine, Taipei 10051, Taiwan; Department of Otolaryngology, National Taiwan University Hospital Hsin-Chu Branch, Hsinchu 302058, Taiwan.
| |
Collapse
|
10
|
Zeng YY, Gu Q, Li D, Li AX, Liu RM, Liang JY, Liu JY. Immunocyte membrane-derived biomimetic nano-drug delivery system: a pioneering platform for tumour immunotherapy. Acta Pharmacol Sin 2024; 45:2455-2473. [PMID: 39085407 PMCID: PMC11579519 DOI: 10.1038/s41401-024-01355-z] [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: 05/16/2024] [Accepted: 07/03/2024] [Indexed: 08/02/2024]
Abstract
Tumor immunotherapy characterized by its high specificity and minimal side effects has achieved revolutionary progress in the field of cancer treatment. However, the complex mechanisms of tumor immune microenvironment (TIME) and the individual variability of patients' immune system still present significant challenges to its clinical application. Immunocyte membrane-coated nanocarrier systems, as an innovative biomimetic drug delivery platform, exhibit remarkable advantages in tumor immunotherapy due to their high targeting capability, good biocompatibility and low immunogenicity. In this review we summarize the latest research advances in biomimetic delivery systems based on immune cells for tumor immunotherapy. We outline the existing methods of tumor immunotherapy including immune checkpoint therapy, adoptive cell transfer therapy and cancer vaccines etc. with a focus on the application of various immunocyte membranes in tumor immunotherapy and their prospects and challenges in drug delivery and immune modulation. We look forward to further exploring the application of biomimetic delivery systems based on immunocyte membrane-coated nanoparticles, aiming to provide a new framework for the clinical treatment of tumor immunity.
Collapse
Affiliation(s)
- Yuan-Ye Zeng
- School of Pharmacy, Fudan University, Shanghai, 201203, China
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Qing Gu
- Department of Pharmacy, Jingan District Zhabei Central Hospital, Shanghai, 200070, China
| | - Dan Li
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Ai-Xue Li
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Rong-Mei Liu
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Jian-Ying Liang
- School of Pharmacy, Fudan University, Shanghai, 201203, China.
| | - Ji-Yong Liu
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
11
|
Fernando V, Zheng X, Sharma V, Sweef O, Choi ES, Furuta S. Reprogramming of breast tumor-associated macrophages with modulation of arginine metabolism. Life Sci Alliance 2024; 7:e202302339. [PMID: 39191486 PMCID: PMC11350068 DOI: 10.26508/lsa.202302339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 08/16/2024] [Accepted: 08/19/2024] [Indexed: 08/29/2024] Open
Abstract
HER2+ breast tumors have abundant immune-suppressive cells, including M2-type tumor-associated macrophages (TAMs). Although TAMs consist of the immune-stimulatory M1 type and immune-suppressive M2 type, the M1/M2-TAM ratio is reduced in immune-suppressive tumors, contributing to their immunotherapy refractoriness. M1- versus M2-TAM formation depends on differential arginine metabolism, where M1-TAMs convert arginine to nitric oxide (NO) and M2-TAMs convert arginine to polyamines (PAs). We hypothesize that such distinct arginine metabolism in M1- versus M2-TAMs is attributed to different availability of BH4 (NO synthase cofactor) and that its replenishment would reprogram M2-TAMs to M1-TAMs. Recently, we reported that sepiapterin (SEP), the endogenous BH4 precursor, elevates the expression of M1-TAM markers within HER2+ tumors. Here, we show that SEP restores BH4 levels in M2-like macrophages, which then redirects arginine metabolism to NO synthesis and converts M2 type to M1 type. The reprogrammed macrophages exhibit full-fledged capabilities of antigen presentation and induction of effector T cells to trigger immunogenic cell death of HER2+ cancer cells. This study substantiates the utility of SEP in the metabolic shift of the HER2+ breast tumor microenvironment as a novel immunotherapeutic strategy.
Collapse
Affiliation(s)
- Veani Fernando
- Department of Cell & Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus, Toledo, OH, USA
- Division of Rheumatology, University of Colorado, Anschutz Medical Campus Barbara Davis Center, Aurora, CO, USA
| | - Xunzhen Zheng
- Department of Cell & Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus, Toledo, OH, USA
| | - Vandana Sharma
- Department of Cell & Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus, Toledo, OH, USA
- Department of Zoology and Physiology, University of Wyoming, Laramie, WY, USA
| | - Osama Sweef
- MetroHealth Medical Center, Case Western Reserve University School of Medicine, Case Comprehensive Cancer Center, Cleveland, OH, USA
| | - Eun-Seok Choi
- MetroHealth Medical Center, Case Western Reserve University School of Medicine, Case Comprehensive Cancer Center, Cleveland, OH, USA
| | - Saori Furuta
- Department of Cell & Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus, Toledo, OH, USA
- MetroHealth Medical Center, Case Western Reserve University School of Medicine, Case Comprehensive Cancer Center, Cleveland, OH, USA
| |
Collapse
|
12
|
Zong YH, Cao JF, Zhao Y, Gao M, Chen WL, Wu M, Xu X, Xu ZY, Zhang XQ, Tang JZ, Liu Y, Hu XS, Wang SQ, Zhang X. Mechanism of Lian Hua Qing Wen capsules regulates the inflammatory response caused by M 1 macrophage based on cellular experiments and computer simulations. Acta Trop 2024; 257:107320. [PMID: 39002739 DOI: 10.1016/j.actatropica.2024.107320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 07/01/2024] [Accepted: 07/04/2024] [Indexed: 07/15/2024]
Abstract
PURPOSE The polarization of macrophages with the resulting inflammatory response play a crucial part in tissue and organ damage due to inflammatory. Study has proved Lian Hua Qing Wen capsules (LHQW) can reduce activation of inflammatory response and damage of tissue derived from the inflammatory reactions. However, the mechanism of LHQW regulates the macrophage-induced inflammatory response is unclear. Therefore, we investigated the mechanism of LHQW regulated the inflammatory response of M1 macrophages by cellular experiments and computer simulations. METHODS This study has analysed the targets and mechanisms of macrophage regulating inflammatory response at gene and protein levels through bioinformatics. The monomeric components of LHQW were analyzed by High Performance Liquid Chromatography (HPLC). We established the in vitro cell model by M1 macrophages (Induction of THP-1 cells into M1 macrophages). RT-qPCR and immunofluorescence were used to detect changes in gene and protein levels of key targets after LHQW treatment. Computer simulations were utilized to verify the binding stability of monomeric components and protein targets. RESULTS Macrophages had 140,690 gene targets, inflammatory response had 12,192 gene targets, intersection gene targets were 11,772. Key monomeric components (including: Pinocembrin, Fargesone-A, Nodakenin and Bowdichione) of LHQW were screened by HPLC. The results of cellular experiments indicated that LHQW could significantly reduce the mRNA expression of CCR5, CSF2, IFNG and TNF, thereby alleviating the inflammatory response caused by M1 macrophage. The computer simulations further validated the binding stability and conformation of key monomeric components and key protein targets, and IFNG/Nodakenin was able to form the most stable binding conformation for its action. CONCLUSION In this study, the mechanism of LHQW inhibits the polarization of macrophages and the resulting inflammatory response was investigated by computer simulations and cellular experiments. We found that LHQW may not only reduce cell damage and death by acting on TNF and CCR5, but also inhibit the immune recognition process and inflammatory response by regulating CSF2 and IFNG to prevent polarization of macrophages. Therefore, these results suggested that LHQW may act through multiple targets to inhibit the polarization of macrophages and the resulting inflammatory response.
Collapse
Affiliation(s)
| | - Jun-Feng Cao
- College of Medicine, Southwest Jiaotong University, Chengdu, PR China
| | | | - Miao Gao
- Chengdu Medical College, Chengdu, PR China
| | | | - Mei Wu
- Chengdu Medical College, Chengdu, PR China
| | - Xiang Xu
- Chengdu Medical College, Chengdu, PR China
| | | | | | | | - Yulin Liu
- Chengdu Medical College, Chengdu, PR China
| | | | | | - Xiao Zhang
- Chengdu Medical College, Chengdu, PR China.
| |
Collapse
|
13
|
Zhou W, Yang F, Zhang X. Roles of M1 Macrophages and Their Extracellular Vesicles in Cancer Therapy. Cells 2024; 13:1428. [PMID: 39273000 PMCID: PMC11394047 DOI: 10.3390/cells13171428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 08/21/2024] [Accepted: 08/22/2024] [Indexed: 09/15/2024] Open
Abstract
Tumor-associated macrophages (TAMs) are inflammatory cells that are important components of the tumor microenvironment. TAMs are functionally heterogeneous and divided into two main subpopulations with distinct and opposite functions: M1 and M2 macrophages. The secretory function of TAMs is essential for combating infections, regulating immune responses, and promoting tissue repair. Extracellular vesicles (EVs) are nanovesicles that are secreted by cells. They play a crucial role in mediating intercellular information transfer between cells. EVs can be secreted by almost all types of cells, and they contain proteins, microRNAs, mRNAs, and even long non-coding RNAs (lncRNAs) that have been retained from the parental cell through the process of biogenesis. EVs can influence the function and behavior of target cells by delivering their contents, thus reflecting, to some extent, the characteristics of their parental cells. Here, we provide an overview of the role of M1 macrophages and their EVs in cancer therapy by exploring the impact of M1 macrophage-derived EVs (M1-EVs) on tumors by transferring small microRNAs. Additionally, we discuss the potential of M1-EVs as drug carriers and the possibility of reprogramming M2 macrophages into M1 macrophages for disease treatment. We propose that M1-EVs play a crucial role in cancer therapy by transferring microRNAs and loading them with drugs. Reprogramming M2 macrophages into M1 macrophages holds great promise in the treatment of cancers.
Collapse
Affiliation(s)
| | | | - Xiuzhen Zhang
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255049, China (F.Y.)
| |
Collapse
|
14
|
Chen T, Ma W, Wang X, Ye Q, Hou X, Wang Y, Jiang C, Meng X, Sun Y, Cai J. Insights of immune cell heterogeneity, tumor-initiated subtype transformation, drug resistance, treatment and detecting technologies in glioma microenvironment. J Adv Res 2024:S2090-1232(24)00315-1. [PMID: 39097088 DOI: 10.1016/j.jare.2024.07.033] [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: 04/07/2024] [Revised: 06/30/2024] [Accepted: 07/29/2024] [Indexed: 08/05/2024] Open
Abstract
BACKGROUND With the gradual understanding of glioma development and the immune microenvironment, many immune cells have been discovered. Despite the growing comprehension of immune cell functions and the clinical application of immunotherapy, the precise roles and characteristics of immune cell subtypes, how glioma induces subtype transformation of immune cells and its impact on glioma progression have yet to be understood. AIM OF THE REVIEW In this review, we comprehensively center on the four major immune cells within the glioma microenvironment, particularly neutrophils, macrophages, lymphocytes, myeloid-derived suppressor cells (MDSCs), and other significant immune cells. We discuss (1) immune cell subtype markers, (2) glioma-induced immune cell subtype transformation, (3) the mechanisms of each subtype influencing chemotherapy resistance, (4) therapies targeting immune cells, and (5) immune cell-associated single-cell sequencing. Eventually, we identified the characteristics of immune cell subtypes in glioma, comprehensively summarized the exact mechanism of glioma-induced immune cell subtype transformation, and concluded the progress of single-cell sequencing in exploring immune cell subtypes in glioma. KEY SCIENTIFIC CONCEPTS OF REVIEW In conclusion, we have analyzed the mechanism of chemotherapy resistance detailly, and have discovered prospective immunotherapy targets, excavating the potential of novel immunotherapies approach that synergistically combines radiotherapy, chemotherapy, and surgery, thereby paving the way for improved immunotherapeutic strategies against glioma and enhanced patient outcomes.
Collapse
Affiliation(s)
- Tongzheng Chen
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wenbin Ma
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xin Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qile Ye
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xintong Hou
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yiwei Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chuanlu Jiang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China; The Six Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiangqi Meng
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Ying Sun
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Jinquan Cai
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
15
|
Zhang W, Wang M, Ji C, Liu X, Gu B, Dong T. Macrophage polarization in the tumor microenvironment: Emerging roles and therapeutic potentials. Biomed Pharmacother 2024; 177:116930. [PMID: 38878638 DOI: 10.1016/j.biopha.2024.116930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 05/31/2024] [Accepted: 06/09/2024] [Indexed: 07/28/2024] Open
Abstract
The tumor microenvironment (TME) is a combination of tumor cells and indigenous host stroma, which consists of tumor-infiltrating immune cells, endothelial cells, fibroblasts, pericytes, and non-cellular elements. Tumor-associated macrophages (TAMs) represent the major tumor-infiltrating immune cell type and are generally polarized into two functionally contradictory subtypes, namely classical activated M1 macrophages and alternatively activated M2 macrophages. Macrophage polarization refers to how macrophages are activated at a given time and space. The interplay between the TME and macrophage polarization can influence tumor initiation and progression, making TAM a potential target for cancer therapy. Here, we review the latest investigations on factors orchestrating macrophage polarization in the TME, how macrophage polarization affects tumor progression, and the perspectives in modulating macrophage polarization for cancer immunotherapy.
Collapse
Affiliation(s)
- Wenru Zhang
- Department of Natural Products Chemistry, Key Laboratory of Natural Products & Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Mengmeng Wang
- Department of Natural Products Chemistry, Key Laboratory of Natural Products & Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Chonghao Ji
- Department of Implantology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Xiaohui Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 2A Nanwei Road, Xicheng District, Beijing 100050, China
| | - Bowen Gu
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, United States.
| | - Ting Dong
- Department of Natural Products Chemistry, Key Laboratory of Natural Products & Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China.
| |
Collapse
|
16
|
Zhao R, Sukocheva O, Tse E, Neganova M, Aleksandrova Y, Zheng Y, Gu H, Zhao D, Madhunapantula SV, Zhu X, Liu J, Fan R. Cuproptosis, the novel type of oxidation-induced cell death in thoracic cancers: can it enhance the success of immunotherapy? Cell Commun Signal 2024; 22:379. [PMID: 39068453 PMCID: PMC11282696 DOI: 10.1186/s12964-024-01743-2] [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: 02/26/2024] [Accepted: 07/08/2024] [Indexed: 07/30/2024] Open
Abstract
Copper is an important metal micronutrient, required for the balanced growth and normal physiological functions of human organism. Copper-related toxicity and dysbalanced metabolism were associated with the disruption of intracellular respiration and the development of various diseases, including cancer. Notably, copper-induced cell death was defined as cuproptosis which was also observed in malignant cells, representing an attractive anti-cancer instrument. Excess of intracellular copper leads to the aggregation of lipoylation proteins and toxic stress, ultimately resulting in the activation of cell death. Differential expression of cuproptosis-related genes was detected in normal and malignant tissues. Cuproptosis-related genes were also linked to the regulation of oxidative stress, immune cell responses, and composition of tumor microenvironment. Activation of cuproptosis was associated with increased expression of redox-metabolism-regulating genes, such as ferredoxin 1 (FDX1), lipoic acid synthetase (LIAS), lipoyltransferase 1 (LIPT1), dihydrolipoamide dehydrogenase (DLD), drolipoamide S-acetyltransferase (DLAT), pyruvate dehydrogenase E1 subunit alpha 1 (PDHA1), and pyruvate dehydrogenase E1 subunit beta (PDHB)). Accordingly, copper-activated network was suggested as an attractive target in cancer therapy. Mechanisms of cuproptosis and regulation of cuproptosis-related genes in different cancers and tumor microenvironment are discussed in this study. The analysis of current findings indicates that therapeutic regulation of copper signaling, and activation of cuproptosis-related targets may provide an effective tool for the improvement of immunotherapy regimens.
Collapse
Affiliation(s)
- Ruiwen Zhao
- The Department of Radiation Oncology & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Olga Sukocheva
- Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Port Rd, Adelaide, SA, 5000, Australia.
| | - Edmund Tse
- Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Port Rd, Adelaide, SA, 5000, Australia
| | - Margarita Neganova
- Nesmeyanov Institute of Organoelement Compounds, Russian Academy of Sciences, Moscow, 119991, Russia
| | - Yulia Aleksandrova
- Nesmeyanov Institute of Organoelement Compounds, Russian Academy of Sciences, Moscow, 119991, Russia
| | - Yufei Zheng
- The Department of Radiation Oncology & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Hao Gu
- The Department of Radiation Oncology & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Deyao Zhao
- The Department of Radiation Oncology & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - SabbaRao V Madhunapantula
- Special Interest Group in Cancer Biology and Cancer Stem Cells (SIG-CBCSC), Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education & Research, Mysuru, Karnataka, 570015, India
| | - Xiaorong Zhu
- The Department of Radiation Oncology & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Junqi Liu
- The Department of Radiation Oncology & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Ruitai Fan
- The Department of Radiation Oncology & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
17
|
Wang H, Yang L, Qiu Q, Rao Q, Liu L, Cui Y, Zhang L, Ma Y, Jin X, Yang D, Qi S, Wang K, Li Y, Zhang X, Zhao M. Exploring the Health Benefits of Boletus aereus Polysaccharides: Extraction, Structural Characterization, and Antiproliferative Properties against Non-Hodgkin's Lymphomas (NHLs). JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:16334-16346. [PMID: 38994810 DOI: 10.1021/acs.jafc.4c03945] [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: 07/13/2024]
Abstract
Boletus aereus Fr. ex Bull. stands out as a delectable edible mushroom with high nutritional and medicinal values, featuring polysaccharides as its primary nutrient composition. In our continuous exploration of its beneficial substances, a novel polysaccharide (BAPN-1) with a molecular weight of 2279 kDa was prepared. It was identified as a glucan with a backbone composed of the residues →4)-α-Glcp-(1→ and →4,6)-α-Glcp-(1→ connected in a proportion of 5:1 and a β-Glcp-(1→ side residue attached at C6 of the →4,6)-α-Glcp-(1→ residue. Biologically, BAPN-1 exhibited broad-spectrum antiproliferative activities against various NHL cells, including HuT-78, OCI-LY1, OCI-LY18, Jurkat, RL, and Karpas-299, with IC50 values of 0.73, 1.21, 3.18, 1.52, 3.34, and 4.25 mg/mL, respectively. Additionally, BAPN-1 significantly induced cell cycle arrest in the G2/M phase and caused apoptosis of NHL cells. Mechanistically, bulk RNA sequencing and Western blot analysis revealed that BAPN-1 could upregulate cyclin B1 and enhance cleaved caspase-9 expression through the inhibition of FGFR3 and RAF-MEK-ERK signaling pathways. This work supports the improved utilization of B. aereus in high-value health products.
Collapse
Affiliation(s)
- Haidi Wang
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital, Sichuan University, Chengdu 610041, China
- Faculty of Chemistry and Chemical Engineering, Yunnan Normal University, Kunming 650500, China
| | - Linyu Yang
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qiang Qiu
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qianru Rao
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital, Sichuan University, Chengdu 610041, China
| | - Li Liu
- Faculty of Chemistry and Chemical Engineering, Yunnan Normal University, Kunming 650500, China
| | - Yuchen Cui
- Faculty of Chemistry and Chemical Engineering, Yunnan Normal University, Kunming 650500, China
| | - Liang Zhang
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China
| | - Yucheng Ma
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xi Jin
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital, Sichuan University, Chengdu 610041, China
| | - Dongxue Yang
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shiqian Qi
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital, Sichuan University, Chengdu 610041, China
| | - Kunjie Wang
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yong Li
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Xufeng Zhang
- Faculty of Chemistry and Chemical Engineering, Yunnan Normal University, Kunming 650500, China
| | - Min Zhao
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
18
|
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
|
19
|
Kang Y, Yan J, Han X, Wang X, Wang Y, Song P, Su X, Rauf A, Jin X, Pu F, Zhang H. Construction of Hierarchically Biomimetic Iron Oxide Nanosystems for Macrophage Repolarization-Promoted Immune Checkpoint Blockade of Cancer Immunotherapy. ACS APPLIED MATERIALS & INTERFACES 2024; 16:36131-36141. [PMID: 38979627 DOI: 10.1021/acsami.4c06415] [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: 07/10/2024]
Abstract
Cancer immunotherapy is developing as the mainstream strategy for treatment of cancer. However, the interaction between the programmed cell death protein-1 (PD-1) and the programmed death ligand 1 (PD-L1) restricts T cell proliferation, resulting in the immune escape of tumor cells. Recently, immune checkpoint inhibitor therapy has achieved clinical success in tumor treatment through blocking the PD-1/PD-L1 checkpoint pathway. However, the presence of M2 tumor-associated macrophages (TAMs) in the tumor microenvironment (TME) will inhibit antitumor immune responses and facilitate tumor growth, which can weaken the effectiveness of immune checkpoint inhibitor therapy. The repolarization of M2 TAMs into M1 TAMs can induce the immune response to secrete proinflammatory factors and active T cells to attack tumor cells. Herein, hollow iron oxide (Fe3O4) nanoparticles (NPs) were prepared for reprogramming M2 TAMs into M1 TAMs. BMS-202, a small-molecule PD-1/PD-L1 inhibitor that has a lower price, higher stability, lower immunogenicity, and higher tumor penetration ability compared with antibodies, was loaded together with pH-sensitive NaHCO3 inside hollow Fe3O4 NPs, followed by wrapping with macrophage membranes. The formed biomimetic FBN@M could produce gaseous carbon dioxide (CO2) from NaHCO3 in response to the acidic TME, breaking up the macrophage membranes to release BMS-202. A series of in vitro and in vivo assessments revealed that FBN@M could reprogram M2 TAMs into M1 TAMs and block the PD-1/PD-L1 pathway, which eventually induced T cell activation and the secretion of TNF-α and IFN-γ to kill the tumor cells. FBN@M has shown a significant immunotherapeutic efficacy for tumor treatment.
Collapse
Affiliation(s)
- Yaqing Kang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Jiao Yan
- School of Biomedical Engineering & The First Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Xiaoqing Han
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Xingbo Wang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Yanjing Wang
- School of Biomedical Engineering & The First Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Panpan Song
- School of Biomedical Engineering & The First Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Xiaochen Su
- Second Inpatient Area of Urology Department, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Abdur Rauf
- Department of Chemistry, University of Swabi, Anbar 23430, Pakistan
| | - Xuefei Jin
- Second Inpatient Area of Urology Department, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Fang Pu
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Haiyuan Zhang
- School of Biomedical Engineering & The First Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| |
Collapse
|
20
|
Herreros-Cabello A, Del Moral-Salmoral J, Morato E, Marina A, Barrocal B, Fresno M, Gironès N. Quantitative Proteomic Analysis of Macrophages Infected with Trypanosoma cruzi Reveals Different Responses Dependent on the SLAMF1 Receptor and the Parasite Strain. Int J Mol Sci 2024; 25:7493. [PMID: 39000601 PMCID: PMC11242706 DOI: 10.3390/ijms25137493] [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: 06/05/2024] [Revised: 06/29/2024] [Accepted: 07/04/2024] [Indexed: 07/16/2024] Open
Abstract
Chagas disease is caused by the intracellular protozoan parasite Trypanosoma cruzi. This disease affects mainly rural areas in Central and South America, where the insect vector is endemic. However, this disease has become a world health problem since migration has spread it to other continents. It is a complex disease with many reservoirs and vectors and high genetic variability. One of the host proteins involved in the pathogenesis is SLAMF1. This immune receptor acts during the infection of macrophages controlling parasite replication and thus affecting survival in mice but in a parasite strain-dependent manner. Therefore, we studied the role of SLAMF1 by quantitative proteomics in a macrophage in vitro infection and the different responses between Y and VFRA strains of Trypanosoma cruzi. We detected different significant up- or downregulated proteins involved in immune regulation processes, which are SLAMF1 and/or strain-dependent. Furthermore, independently of SLAMF1, this parasite induces different responses in macrophages to counteract the infection and kill the parasite, such as type I and II IFN responses, NLRP3 inflammasome activation, IL-18 production, TLR7 and TLR9 activation specifically with the Y strain, and IL-11 signaling specifically with the VFRA strain. These results have opened new research fields to elucidate the concrete role of SLAMF1 and discover new potential therapeutic approaches for Chagas disease.
Collapse
Affiliation(s)
- Alfonso Herreros-Cabello
- Departamento de Biología Molecular, Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain
- Centro de Biología Molecular Severo Ochoa (CBMSO), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28049 Madrid, Spain
| | - Javier Del Moral-Salmoral
- Departamento de Biología Molecular, Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain
- Centro de Biología Molecular Severo Ochoa (CBMSO), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28049 Madrid, Spain
| | - Esperanza Morato
- Unidad de Proteómica, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), 28049 Madrid, Spain
| | - Anabel Marina
- Unidad de Proteómica, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), 28049 Madrid, Spain
- Unidad de Técnicas Bioanalíticas (BAT), Instituto de Investigación de Ciencias de la Alimentación (CIAL), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28049 Madrid, Spain
| | - Beatriz Barrocal
- Centro de Biología Molecular Severo Ochoa (CBMSO), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28049 Madrid, Spain
| | - Manuel Fresno
- Departamento de Biología Molecular, Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain
- Centro de Biología Molecular Severo Ochoa (CBMSO), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28049 Madrid, Spain
- Instituto Universitario de Biología Molecular, Universidad Autónoma de Madrid (IUBM-UAM), 28049 Madrid, Spain
- Instituto de Investigación Sanitaria, Hospital Universitario de La Princesa, 28006 Madrid, Spain
| | - Núria Gironès
- Departamento de Biología Molecular, Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain
- Centro de Biología Molecular Severo Ochoa (CBMSO), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28049 Madrid, Spain
- Instituto Universitario de Biología Molecular, Universidad Autónoma de Madrid (IUBM-UAM), 28049 Madrid, Spain
- Instituto de Investigación Sanitaria, Hospital Universitario de La Princesa, 28006 Madrid, Spain
| |
Collapse
|
21
|
Zhang J, Ma Y. Luteolin as a potential therapeutic candidate for lung cancer: Emerging preclinical evidence. Biomed Pharmacother 2024; 176:116909. [PMID: 38852513 DOI: 10.1016/j.biopha.2024.116909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/04/2024] [Accepted: 06/06/2024] [Indexed: 06/11/2024] Open
Abstract
Lung cancer is a prevalent malignant tumor and a leading cause of cancer-related fatalities globally. However, current treatments all have limitations. Therefore, there is an urgent need to identify a readily available therapeutic agent to counteract lung cancer development and progression. Luteolin is a flavonoid derived from vegetables and herbs that possesses preventive and therapeutic effects on various cancers. With the goal of providing new directions for the treatment of lung cancer, we review here the recent findings on luteolin so as to provide new ideas for the development of new anti-lung cancer drugs. The search focused on studies published between January 1995 and January 2024 that explored the use of luteolin in lung cancer. A comprehensive literature search was conducted in the SCOPUS, Google Scholar, PubMed, and Web of Science databases using the keywords "luteolin" and "lung cancer." By collecting previous literature, we found that luteolin has multiple mechanisms of therapeutic effects, including promotion of apoptosis in lung cancer cells; inhibition of tumor cell proliferation, invasion and metastasis; and modulation of immune responses. In addition, it can be used as an adjuvant to radio-chemotherapy and helps to ameliorate cancer complications. This review summarizes the structure, natural sources, physicochemical properties and pharmacokinetics of luteolin, and focuses on the anti-lung cancer mechanism of luteolin, so as to provide new ideas for the development of new anti-lung cancer drugs.
Collapse
Affiliation(s)
- Jin Zhang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, Liaoning 110004, PR China
| | - Yue Ma
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, Liaoning 110004, PR China.
| |
Collapse
|
22
|
Xu M, Li S. The opportunities and challenges of using PD-1/PD-L1 inhibitors for leukemia treatment. Cancer Lett 2024; 593:216969. [PMID: 38768681 DOI: 10.1016/j.canlet.2024.216969] [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: 12/14/2023] [Revised: 05/14/2024] [Accepted: 05/15/2024] [Indexed: 05/22/2024]
Abstract
Leukemia poses a significant clinical challenge due to its swift onset, rapid progression, and treatment-related complications. Tumor immune evasion, facilitated by immune checkpoints like programmed death receptor 1/programmed death receptor ligand 1 (PD-1/PD-L1), plays a critical role in leukemia pathogenesis and progression. In this review, we summarized the research progress and therapeutic potential of PD-L1 in leukemia, focusing on targeted therapy and immunotherapy. Recent clinical trials have demonstrated promising outcomes with PD-L1 inhibitors, highlighting their role in enhancing treatment efficacy. This review discusses the implications of PD-L1 expression levels on treatment response and long-term survival rates in leukemia patients. Furthermore, we address the challenges and opportunities in immunotherapy, emphasizing the need for personalized approaches and combination therapies to optimize PD-L1 inhibition in leukemia management. Future research prospects include exploring novel treatment strategies and addressing immune-related adverse events to improve clinical outcomes in leukemia. Overall, this review provides valuable insights into the role of PD-L1 in leukemia and its potential as a therapeutic target in the evolving landscape of leukemia treatment.
Collapse
Affiliation(s)
- Mengdan Xu
- Department of Breast Cancer, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, 110042, China; Institute of Cancer Medicine, Dalian University of Technology, No.2 Linggong Road, Ganjingzi District, Dalian, 116024, Liaoning Province, China
| | - Shenglong Li
- Second Ward of Bone and Soft Tissue Tumor Surgery, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, 110042, China; The Liaoning Provincial Key Laboratory of Interdisciplinary Research on Gastrointestinal Tumor Combining Medicine with Engineering, China; Institute of Cancer Medicine, Dalian University of Technology, No.2 Linggong Road, Ganjingzi District, Dalian, 116024, Liaoning Province, China.
| |
Collapse
|
23
|
Li H, Wang S, Yang Z, Meng X, Niu M. Nanomaterials modulate tumor-associated macrophages for the treatment of digestive system tumors. Bioact Mater 2024; 36:376-412. [PMID: 38544737 PMCID: PMC10965438 DOI: 10.1016/j.bioactmat.2024.03.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 02/25/2024] [Accepted: 03/03/2024] [Indexed: 11/25/2024] Open
Abstract
The treatment of digestive system tumors presents challenges, particularly in immunotherapy, owing to the advanced immune tolerance of the digestive system. Nanomaterials have emerged as a promising approach for addressing these challenges. They provide targeted drug delivery, enhanced permeability, high bioavailability, and low toxicity. Additionally, nanomaterials target immunosuppressive cells and reshape the tumor immune microenvironment (TIME). Among the various cells in the TIME, tumor-associated macrophages (TAMs) are the most abundant and play a crucial role in tumor progression. Therefore, investigating the modulation of TAMs by nanomaterials for the treatment of digestive system tumors is of great significance. Here, we present a comprehensive review of the utilization of nanomaterials to modulate TAMs for the treatment of gastric cancer, colorectal cancer, hepatocellular carcinoma, and pancreatic cancer. We also investigated the underlying mechanisms by which nanomaterials modulate TAMs to treat tumors in the digestive system. Furthermore, this review summarizes the role of macrophage-derived nanomaterials in the treatment of digestive system tumors. Overall, this research offers valuable insights into the development of nanomaterials tailored for the treatment of digestive system tumors.
Collapse
Affiliation(s)
- Hao Li
- Department of Interventional Radiology, First Hospital of China Medical University, Shenyang, China
| | - Shuai Wang
- Department of Interventional Radiology, First Hospital of China Medical University, Shenyang, China
| | - Zhengqiang Yang
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xianwei Meng
- Laboratory of Controllable Preparation and Application of Nanomaterials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, China
| | - Meng Niu
- China Medical University, Shenyang, China
| |
Collapse
|
24
|
Zeng X, Wang TW, Yamaguchi K, Hatakeyama S, Yamazaki S, Shimizu E, Imoto S, Furukawa Y, Johmura Y, Nakanishi M. M2 macrophage-derived TGF-β induces age-associated loss of adipogenesis through progenitor cell senescence. Mol Metab 2024; 84:101943. [PMID: 38657734 PMCID: PMC11079528 DOI: 10.1016/j.molmet.2024.101943] [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: 11/24/2023] [Revised: 04/04/2024] [Accepted: 04/16/2024] [Indexed: 04/26/2024] Open
Abstract
OBJECTIVES Adipose tissue is an endocrine and energy storage organ composed of several different cell types, including mature adipocytes, stromal cells, endothelial cells, and a variety of immune cells. Adipose tissue aging contributes to the pathogenesis of metabolic dysfunction and is likely induced by crosstalk between adipose progenitor cells (APCs) and immune cells, but the underlying molecular mechanisms remain largely unknown. In this study, we revealed the biological role of p16high senescent APCs, and investigated the crosstalk between each cell type in the aged white adipose tissue. METHODS We performed the single-cell RNA sequencing (scRNA-seq) analysis on the p16high adipose cells sorted from aged p16-CreERT2/Rosa26-LSL-tdTomato mice. We also performed the time serial analysis on the age-dependent bulk RNA-seq datasets of human and mouse white adipose tissues to infer the transcriptome alteration of adipogenic potential within aging. RESULTS We show that M2 macrophage-derived TGF-β induces APCs senescence which impairs adipogenesis in vivo. p16high senescent APCs increase with age and show loss of adipogenic potential. The ligand-receptor interaction analysis reveals that M2 macrophages are the donors for TGF-β and the senescent APCs are the recipients. Indeed, treatment of APCs with TGF-β1 induces senescent phenotypes through mitochondrial ROS-mediated DNA damage in vitro. TGF-β1 injection into gonadal white adipose tissue (gWAT) suppresses adipogenic potential and induces fibrotic genes as well as p16 in APCs. A gWAT atrophy is observed in cancer cachexia by APCs senescence, whose induction appeared to be independent of TGF-β induction. CONCLUSIONS Our results suggest that M2 macrophage-derived TGF-β induces age-related lipodystrophy by APCs senescence. The TGF-β treatment induced DNA damage, mitochondrial ROS, and finally cellular senescence in APCs.
Collapse
Affiliation(s)
- Xinyi Zeng
- Division of Cancer Cell Biology, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Teh-Wei Wang
- Division of Cancer Cell Biology, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan.
| | - Kiyoshi Yamaguchi
- Division of Clinical Genome Research, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Seira Hatakeyama
- Division of Clinical Genome Research, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Satoshi Yamazaki
- Division of Stem Cell Biology, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Eigo Shimizu
- Division of Health Medical Intelligence, Human Genome Center, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Seiya Imoto
- Division of Health Medical Intelligence, Human Genome Center, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Yoichi Furukawa
- Division of Clinical Genome Research, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Yoshikazu Johmura
- Division of Cancer and Senescence Biology, Cancer Research Institute, Institute for Frontier Science Initiative, Kanazawa University, Kanazawa, Japan
| | - Makoto Nakanishi
- Division of Cancer Cell Biology, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan.
| |
Collapse
|
25
|
Sun G, Liu C, Lu Z, Zhang J, Cao H, Huang T, Dai M, Liu H, Feng T, Tang W, Xia Y. Metabolomics reveals ascorbic acid inhibits ferroptosis in hepatocytes and boosts the effectiveness of anti-PD1 immunotherapy in hepatocellular carcinoma. Cancer Cell Int 2024; 24:192. [PMID: 38822322 PMCID: PMC11143590 DOI: 10.1186/s12935-024-03342-0] [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: 02/13/2023] [Accepted: 04/23/2024] [Indexed: 06/02/2024] Open
Abstract
BACKGROUND Immunotherapy combined with molecular targeted therapy is increasingly popular in patients with advanced hepatocellular carcinoma (HCC). However, immune-related adverse events(irAEs) brought on by immunotherapy increase the likelihood of side effects, thus it is important to look into ways to address this issue. METHODS Different metabolite patterns were established by analyzing metabolomics data in liver tissue samples from 10 patients(divided into severe and mild liver injury) before and after immuno-targeted therapy. After establishing a subcutaneous tumor model of HCC, the mice were divided into PBS group, ascorbic acid(AA) group, and anti-PD1 + tyrosine kinase inhibitor (TKI) group, anti-PD1 + TKI + AA group. Liver tissue were stained with hematoxylin-eosin staining(HE) and the content of aspartate transaminase (AST) and alanine transaminase(ALT) in blood were determined. The mechanism was confirmed by western blotting, mass cytometry, and other techniques. RESULTS Through metabolomics analysis, AA was significantly reduced in the sample of patients with severe liver injury caused by immuno-targeted therapy compared to patients with mild liver injury. The addition of AA in vivo experiments demonstrated a reduction in liver injury in mice. In the liver tissues of the anti-PD1 + TKI + AA group, the protein expressions of SLC7A11,GPX4 and the level of glutathione(GSH) were found to be higher compared to the anti-PD1 + TKI group. Mass cytometry analysis revealed a significant increase in the CD11b+CD44+ PD-L1+ cell population in the AA group when compared to the PBS group. CONCLUSIONS AA could reduce liver injury by preventing hepatocyte SLC7A11/GPX4 ferroptosis and improve the immunotherapy effect of anti-PD1 by boosting CD11b+CD44+PD-L1+cell population in HCC.
Collapse
Affiliation(s)
- Guoqiang Sun
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key laboratory of Hepatobiliary cancers, Nanjing, Jiangsu, China
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chuan Liu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key laboratory of Hepatobiliary cancers, Nanjing, Jiangsu, China
| | - Zhengqing Lu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key laboratory of Hepatobiliary cancers, Nanjing, Jiangsu, China
| | - Jinyu Zhang
- Central Laboratory, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hengsong Cao
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key laboratory of Hepatobiliary cancers, Nanjing, Jiangsu, China
| | - Tian Huang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key laboratory of Hepatobiliary cancers, Nanjing, Jiangsu, China
| | - Mingrui Dai
- Stomatological college of Nanjing Medical University, Nanjing, China
| | - Hanyuan Liu
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Tingting Feng
- Central Laboratory, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Weiwei Tang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key laboratory of Hepatobiliary cancers, Nanjing, Jiangsu, China.
| | - Yongxiang Xia
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key laboratory of Hepatobiliary cancers, Nanjing, Jiangsu, China.
| |
Collapse
|
26
|
Yu X, Li S, Ke S, Ye C, Wang Q, Wang H, Wang L. CSF2 Impairs Nrf2 Signaling through the Akt/Mtor Pathway in the Development of Bladder Cancer. J Cancer 2024; 15:3242-3253. [PMID: 38817867 PMCID: PMC11134447 DOI: 10.7150/jca.94343] [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: 01/16/2024] [Accepted: 04/02/2024] [Indexed: 06/01/2024] Open
Abstract
Bladder Cancer (BCa) is one of the most common cancers of the urinary system. Colony-stimulating factor 2 (CSF2) is involved in many cancers, but not BCa. We investigated the effect of CSF2 on BCa in this study and the underlying molecular mechanisms. CSF2 mRNA levels in BCa were analyzed using the Cancer Genome Atlas (TCGA) database. Western blot was conducted to verify CSF2 expression in BCa tissue samples and cell lines. The effect of CSF2 on the growth of BCa cells was assessed by CCK8 and colony formation. To determine the migration and invasion capabilities of BCa cells, transwell analysis and wound healing assays were conducted. Next, western blot was used to explore the underlying mechanism. In the end, a xenografted BCa mouse model was established to examine the effects of CSF2 on tumorigenesis in vivo. Results showed that CSF2 mRNA was upregulated in BCa samples. Knocking down CSF2 significantly inhibited the proliferation and tumorigenesis of BCa cells in vitro and in vivo. Mechanism analysis revealed that CSF2 knockdown inhibited the proliferation and invasion of BCa cells via AKT/mTOR signaling. Based on these results, CSF2 promotes the proliferation and tumorigenesis of BCa.
Collapse
Affiliation(s)
- Xi Yu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Shenglan Li
- Department of Radiography, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Shuai Ke
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Chenglin Ye
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Qinghua Wang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Huaxin Wang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Lei Wang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| |
Collapse
|
27
|
Yi L, Gai Y, Chen Z, Tian K, Liu P, Liang H, Xu X, Peng Q, Luo X. Macrophage colony-stimulating factor and its role in the tumor microenvironment: novel therapeutic avenues and mechanistic insights. Front Oncol 2024; 14:1358750. [PMID: 38646440 PMCID: PMC11027505 DOI: 10.3389/fonc.2024.1358750] [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: 12/20/2023] [Accepted: 03/12/2024] [Indexed: 04/23/2024] Open
Abstract
The tumor microenvironment is a complex ecosystem where various cellular and molecular interactions shape the course of cancer progression. Macrophage colony-stimulating factor (M-CSF) plays a pivotal role in this context. This study delves into the biological properties and functions of M-CSF in regulating tumor-associated macrophages (TAMs) and its role in modulating host immune responses. Through the specific binding to its receptor colony-stimulating factor 1 receptor (CSF-1R), M-CSF orchestrates a cascade of downstream signaling pathways to modulate macrophage activation, polarization, and proliferation. Furthermore, M-CSF extends its influence to other immune cell populations, including dendritic cells. Notably, the heightened expression of M-CSF within the tumor microenvironment is often associated with dismal patient prognoses. Therefore, a comprehensive investigation into the roles of M-CSF in tumor growth advances our comprehension of tumor development mechanisms and unveils promising novel strategies and approaches for cancer treatment.
Collapse
Affiliation(s)
- Li Yi
- Medical Technology College of Qiqihar Medical College, Qiqihar, Heilongjiang, China
| | - Yihan Gai
- School of Stomatology, Qiqihar Medical College, Qiqihar, Heilongjiang, China
| | - Zhuo Chen
- Medical Technology College of Qiqihar Medical College, Qiqihar, Heilongjiang, China
| | - Kecan Tian
- Medical Technology College of Qiqihar Medical College, Qiqihar, Heilongjiang, China
| | - Pengfei Liu
- School of Basic Medical Sciences, Qiqihar Medical College, Qiqihar, Heilongjiang, China
| | - Hongrui Liang
- School of Basic Medical Sciences, Qiqihar Medical College, Qiqihar, Heilongjiang, China
| | - Xinyu Xu
- Medical Technology College of Qiqihar Medical College, Qiqihar, Heilongjiang, China
| | - Qiuyi Peng
- School of Basic Medical Sciences, Qiqihar Medical College, Qiqihar, Heilongjiang, China
| | - Xiaoqing Luo
- Medical Technology College of Qiqihar Medical College, Qiqihar, Heilongjiang, China
| |
Collapse
|
28
|
Baş Y, Yilmaz B, Acar SF, Karadağ İ. Programmed Cell Death Ligand 1 Expression in CD163 + Tumor-associated Macrophages in Cancer Gland Rupture Microenvironment. Appl Immunohistochem Mol Morphol 2024; 32:176-182. [PMID: 38314768 DOI: 10.1097/pai.0000000000001186] [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: 09/12/2023] [Accepted: 01/15/2024] [Indexed: 02/07/2024]
Abstract
In this study, we aimed to examine the relationship among cancer gland rupture microenvironment, programmed cell death ligand 1 (PD-L1) expression in CD163 + tumor-associated macrophages (TAMs), and prognosis in colon adenocarcinoma. A total of 122 patients were diagnosed with colon adenocarcinoma between 2010 and 2019. PD-L1 + (clone 22C3) "macrophage scores" in the microenvironment of cancer gland rupture were calculated. The effects of these variables on prognosis were statistically analyzed. CD163 + TAMs were denser in the cancer gland rupture microenvironment. PD-L1 + TAMs were observed in the tumor periphery, and there was a significant difference between the rates of PD-L1 expression in TAMs and survival time (log-rank = 10.46, P = 0.015), clinical stage 2 ( P = 0.038), and primary tumor 3 and primary tumor 4 cases ( P = 0.004, P = 0.013). The risk of mortality was 4.070 times higher in patients with a PD-L1 expression rate of ≥1% in CD163 + TAMs. High PD-L1 expression in CD163 + TAMs is associated with poor overall survival. Therefore, blocking PD-L1 in CD163 + TAMs can be used as a target for immunotherapy.
Collapse
Affiliation(s)
- Yilmaz Baş
- Department of Pathology, Faculty of Medicine
| | | | | | - İbrahim Karadağ
- Department of Oncology, Erol Olçok Education and Research Hospital, Hitit University, Çorum, Turkey
| |
Collapse
|
29
|
Wang H, Wang X, Zhang X, Xu W. The promising role of tumor-associated macrophages in the treatment of cancer. Drug Resist Updat 2024; 73:101041. [PMID: 38198845 DOI: 10.1016/j.drup.2023.101041] [Citation(s) in RCA: 36] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 12/16/2023] [Accepted: 12/28/2023] [Indexed: 01/12/2024]
Abstract
Macrophages are important components of the immune system. Mature macrophages can be recruited to tumor microenvironment that affect tumor cell proliferation, invasion and metastasis, extracellular matrix remodeling, immune suppression, as well as chemotherapy resistance. Classically activated type I macrophages (M1) exhibited marked tumor killing and phagocytosis. Therefore, using macrophages for adoptive cell therapy has attracted attention and become one of the most effective strategies for cancer treatment. Through cytokines and/or chemokines, macrophage can inhibit myeloid cells recruitment, and activate anti-tumor and immune killing functions. Applying macrophages for anti-tumor delivery is one of the most promising approaches for cancer therapy. This review article introduces the role of macrophages in tumor development and drug resistance, and the possible clinical application of targeting macrophages for overcoming drug resistance and enhancing cancer therapeutics, as well as its challenges.
Collapse
Affiliation(s)
- Hongbin Wang
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, PR China; Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, PR China; Department of Surgical Oncology, Harbin Medical University Cancer Hospital, PR China.
| | - Xueying Wang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, PR China; Otolaryngology Major Disease Research Key Laboratory of Hunan Province, PR China
| | - Xin Zhang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, PR China; Otolaryngology Major Disease Research Key Laboratory of Hunan Province, PR China
| | - Wanhai Xu
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, PR China; Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, PR China; Department of Urology, Harbin Medical University Cancer Hospital, PR China.
| |
Collapse
|
30
|
Wang B, Mao Z, Ye J, Jiao X, Zhang T, Wang Q, Han S, Zhang Y, Wang C, Dong T, Cui B. Glycolysis Induced by METTL14 Is Essential for Macrophage Phagocytosis and Phenotype in Cervical Cancer. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:723-736. [PMID: 38197667 PMCID: PMC10828180 DOI: 10.4049/jimmunol.2300339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 10/03/2023] [Indexed: 01/11/2024]
Abstract
N 6-methyladenosine (m6A) is the most abundant mRNA modification in mammals and it plays a vital role in various biological processes. However, the roles of m6A on cervical cancer tumorigenesis, especially macrophages infiltrated in the tumor microenvironment of cervical cancer, are still unclear. We analyzed the abnormal m6A methylation in cervical cancer, using CaSki and THP-1 cell lines, that might influence macrophage polarization and/or function in the tumor microenvironment. In addition, C57BL/6J and BALB/c nude mice were used for validation in vivo. In this study, m6A methylated RNA immunoprecipitation sequencing analysis revealed the m6A profiles in cervical cancer. Then, we discovered that the high expression of METTL14 (methyltransferase 14, N6-adenosine-methyltransferase subunit) in cervical cancer tissues can promote the proportion of programmed cell death protein 1 (PD-1)-positive tumor-associated macrophages, which have an obstacle to devour tumor cells. Functionally, changes of METTL14 in cervical cancer inhibit the recognition and phagocytosis of macrophages to tumor cells. Mechanistically, the abnormality of METTL14 could target the glycolysis of tumors in vivo and vitro. Moreover, lactate acid produced by tumor glycolysis has an important role in the PD-1 expression of tumor-associated macrophages as a proinflammatory and immunosuppressive mediator. In this study, we revealed the effect of glycolysis regulated by METTL14 on the expression of PD-1 and phagocytosis of macrophages, which showed that METTL14 was a potential therapeutic target for treating advanced human cancers.
Collapse
Affiliation(s)
- Bingyu Wang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, People’s Republic of China
| | - Zhonghao Mao
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, People’s Republic of China
| | - Jinwen Ye
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, People’s Republic of China
| | - Xinlin Jiao
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, People’s Republic of China
| | - Teng Zhang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, People’s Republic of China
| | - Qi Wang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, People’s Republic of China
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Shandong First Medical University, Jinan, People’s Republic of China
| | - Sai Han
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, People’s Republic of China
| | - Youzhong Zhang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, People’s Republic of China
| | - Chunling Wang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, People’s Republic of China
| | - Taotao Dong
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, People’s Republic of China
| | - Baoxia Cui
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, People’s Republic of China
| |
Collapse
|
31
|
Liu T, Xu C, Guo J, He Z, Zhang Y, Feng Y. Whole Blood Transcriptome Analysis in Patients with Trigeminal Neuralgia: a Prospective Clinical Study. J Mol Neurosci 2024; 74:16. [PMID: 38300339 DOI: 10.1007/s12031-024-02195-6] [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] [Received: 11/11/2023] [Accepted: 01/28/2024] [Indexed: 02/02/2024]
Abstract
Trigeminal neuralgia (TN) brings a huge burden to patients, without long-term effective treatment. This study aimed to explore the differentially expressed genes (DEGs) and related enrichment pathways in patients with TN. This was a study of transcriptome sequencing and bioinformatics analysis of human samples. Whole blood samples were collected from the TN patients and pain-free controls. RNA was extracted to conduct the RNA-sequencing and the subsequent bioinformatics analysis. DEGs between the two groups were derived. Kyoto encyclopedia of genes and genomes (KEGG) and Gene ontology (GO) was used to find the enrichment pathways of DEGs. Protein protein interaction (PPI) network was used to depict the interaction between DEGs and find the most important gene, hub gene. Compared with the control group, there were 117 up-regulated DEGs and 103 down-regulated DEGs in the whole blood of patients in the TN group. Pathway enrichment analysis showed that DEGs were mainly enriched in the neuroimmune and metabolic pathways. The PPI network demonstrated that colony stimulating factor 2 (CSF2) was the most important hub gene in the whole blood of TN patients. This study shows the expression of the transcriptome in the whole blood samples of TN patients. The neuroimmune responses and key hub gene CSF2 in the whole blood cells play a vital role in the occurrence of TN. Our research provides a theoretical basis for the diagnosis and treatments of TN. This study was registered at clinicaltrials.gov in June 2021 (No. NCT04923399).
Collapse
Affiliation(s)
- Tianyu Liu
- Department of Anesthesiology, Peking University People's Hospital, Xizhimen South Street 11, Beijing, 100044, China
| | - Chao Xu
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jiaqi Guo
- Shanghai Minhang Center for Disease Control and Prevention, Shanghai, China
| | - Zile He
- Department of Anesthesiology, Peking University People's Hospital, Xizhimen South Street 11, Beijing, 100044, China
| | - Yunpeng Zhang
- Department of Anesthesiology, Peking University People's Hospital, Xizhimen South Street 11, Beijing, 100044, China
| | - Yi Feng
- Department of Anesthesiology, Peking University People's Hospital, Xizhimen South Street 11, Beijing, 100044, China.
- Key Laboratory for Neuroscience, Ministry of Education/National Health Commission of China, Peking University, Xueyuan road 38, Beijing, 100191, China.
| |
Collapse
|
32
|
Festekdjian T, Bonavida B. Targeting the Depletion of M2 Macrophages: Implication in Cancer Immunotherapy. Crit Rev Oncog 2024; 29:55-73. [PMID: 38989738 DOI: 10.1615/critrevoncog.2024053580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2024]
Abstract
We have witnessed the emergence of immunotherapy against various cancers that resulted in significant clinical responses and particularly in cancers that were resistant to chemotherapy. These milestones have ignited the development of novel strategies to boost the anti-tumor immune response for immune-suppressed tumors in the tumor microenvironment (TME). Tumor-associated macrophages (TAMs) are the most abundant cells in the TME, and their frequency correlates with poor prognosis. Hence, several approaches have been developed to target TAMs in effort to restore the anti-tumor immune response and inhibit tumor growth and metastasis. One approach discussed herein is targeting TAMs via their depletion. Several methods have been reported for TAMs depletion including micro-RNAs, transcription factors (e.g., PPARγ, KLF4, STAT3, STAT6, NF-κB), chemokines and chemokine receptors, antibodies-mediated blocking the CSF-1/CSF-1R pathway, nanotechnology, and various combination treatments. In addition, various clinical trials are currently examining the targeting of TAMs. Many of these methods also have side effects that need to be monitored and reduced. Future perspectives and directions are discussed.
Collapse
Affiliation(s)
- Talia Festekdjian
- Department of Microbiology, Immunology & Molecular Genetics, David Geffen School of Medicine, Jonsson Comprehensive Cancer Center, University of California at Los Angeles, Los Angeles, CA 90095, USA
| | - Benjamin Bonavida
- Department of Microbiology, Immunology, & Molecular Genetics, David Geffen School of Medicine at UCLA, Johnson Comprehensive Cancer Center, University of California at Los Angeles, Los Angeles, CA 90025-1747, USA
| |
Collapse
|
33
|
Ridnour LA, Cheng RYS, Heinz WF, Pore M, Gonzalez AL, Femino EL, Moffat R, Wink AL, Imtiaz F, Coutinho L, Butcher D, Edmondson EF, Rangel MC, Wong STC, Lipkowitz S, Glynn S, Vitek MP, McVicar DW, Li X, Anderson SK, Paolocci N, Hewitt SM, Ambs S, Billiar TR, Chang JC, Lockett SJ, Wink DA. Spatial analysis of NOS2 and COX2 interaction with T-effector cells reveals immunosuppressive landscapes associated with poor outcome in ER- breast cancer patients. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.21.572867. [PMID: 38187660 PMCID: PMC10769421 DOI: 10.1101/2023.12.21.572867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Multiple immunosuppressive mechanisms exist in the tumor microenvironment that drive poor outcomes and decrease treatment efficacy. The co-expression of NOS2 and COX2 is a strong predictor of poor prognosis in ER- breast cancer and other malignancies. Together, they generate pro-oncogenic signals that drive metastasis, drug resistance, cancer stemness, and immune suppression. Using an ER- breast cancer patient cohort, we found that the spatial expression patterns of NOS2 and COX2 with CD3+CD8+PD1- T effector (Teff) cells formed a tumor immune landscape that correlated with poor outcome. NOS2 was primarily associated with the tumor-immune interface, whereas COX2 was associated with immune desert regions of the tumor lacking Teff cells. A higher ratio of NOS2 or COX2 to Teff was highly correlated with poor outcomes. Spatial analysis revealed that regional clustering of NOS2 and COX2 was associated with stromal-restricted Teff, while only COX2 was predominant in immune deserts. Examination of other immunosuppressive elements, such as PDL1/PD1, Treg, B7H4, and IDO1, revealed that PDL1/PD1, Treg, and IDO1 were primarily associated with restricted Teff, whereas B7H4 and COX2 were found in tumor immune deserts. Regardless of the survival outcome, other leukocytes, such as CD4 T cells and macrophages, were primarily in stromal lymphoid aggregates. Finally, in a 4T1 model, COX2 inhibition led to a massive cell infiltration, thus validating the hypothesis that COX2 is an essential component of the Teff exclusion process and, thus, tumor evasion. Our study indicates that NOS2/COX2 expression plays a central role in tumor immunosuppression. Our findings indicate that new strategies combining clinically available NOS2/COX2 inhibitors with various forms of immune therapy may open a new avenue for the treatment of aggressive ER-breast cancers.
Collapse
Affiliation(s)
- Lisa A Ridnour
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD
| | - Robert Y S Cheng
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD
| | - William F Heinz
- Optical Microscopy and Analysis Laboratory, Frederick National Laboratory for Cancer Research; Leidos Biomedical Research Inc. for the National Cancer Institute, Frederick, MD
| | - Milind Pore
- Imaging Mass Cytometry Frederick National Laboratory for Cancer Research
| | - Ana L Gonzalez
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD
| | - Elise L Femino
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD
| | - Rebecca Moffat
- Optical Microscopy and Analysis Laboratory, Frederick National Laboratory for Cancer Research; Leidos Biomedical Research Inc. for the National Cancer Institute, Frederick, MD
| | - Adelaide L Wink
- Optical Microscopy and Analysis Laboratory, Frederick National Laboratory for Cancer Research; Leidos Biomedical Research Inc. for the National Cancer Institute, Frederick, MD
| | - Fatima Imtiaz
- Optical Microscopy and Analysis Laboratory, Frederick National Laboratory for Cancer Research; Leidos Biomedical Research Inc. for the National Cancer Institute, Frederick, MD
| | - Leandro Coutinho
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD
- Faculdade de Medicina da Universidade de São Paulo and Comprehensive Center for Precision Oncology, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Donna Butcher
- Molecular Histopathology Laboratories, Leidos Biomedical Research Inc. for the National Cancer Institute
| | - Elijah F Edmondson
- Molecular Histopathology Laboratories, Leidos Biomedical Research Inc. for the National Cancer Institute
| | - M Cristina Rangel
- Faculdade de Medicina da Universidade de São Paulo and Comprehensive Center for Precision Oncology, Universidade de São Paulo, São Paulo, SP, Brazil
| | | | | | - Sharon Glynn
- Discipline of Pathology, Lambe Institute for Translational Research, School of Medicine, University of Galway, Galway, Ireland
| | | | - Daniel W McVicar
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD
| | - Xiaoxian Li
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA
| | - Stephen K Anderson
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD
- Basic Science Program, Frederick National Laboratory for Cancer Research
| | - Nazareno Paolocci
- Division of Cardiology, Department of Medicine, Johns Hopkins University, and Department of Biomedical Sciences, University of Padova, Italy
- Laboratory of Pathology CCR, NCI, NIH
| | | | - Stefan Ambs
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Timothy R Billiar
- Mary and Ron Neal Cancer Center, Houston Methodist Hospital and Weill Cornell Medicine, Houston, TX
| | - Jenny C Chang
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD
- Optical Microscopy and Analysis Laboratory, Frederick National Laboratory for Cancer Research; Leidos Biomedical Research Inc. for the National Cancer Institute, Frederick, MD
- Imaging Mass Cytometry Frederick National Laboratory for Cancer Research
- Faculdade de Medicina da Universidade de São Paulo and Comprehensive Center for Precision Oncology, Universidade de São Paulo, São Paulo, SP, Brazil
- Molecular Histopathology Laboratories, Leidos Biomedical Research Inc. for the National Cancer Institute
- Houston Methodist Weill Cornell Medical College, Houston TX
- Women's Malignancies Branch, CCR, NCI, NIH
- Discipline of Pathology, Lambe Institute for Translational Research, School of Medicine, University of Galway, Galway, Ireland
- (Mike Duke)
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA
- Basic Science Program, Frederick National Laboratory for Cancer Research
- Division of Cardiology, Department of Medicine, Johns Hopkins University, and Department of Biomedical Sciences, University of Padova, Italy
- Laboratory of Pathology CCR, NCI, NIH
- Laboratory of Human Carcinogenesis, CCR, NCI, NIH, Bethesda, MD
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA
- Mary and Ron Neal Cancer Center, Houston Methodist Hospital and Weill Cornell Medicine, Houston, TX
| | - Stephen J Lockett
- Optical Microscopy and Analysis Laboratory, Frederick National Laboratory for Cancer Research; Leidos Biomedical Research Inc. for the National Cancer Institute, Frederick, MD
| | - David A Wink
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD
| |
Collapse
|
34
|
Cavallone IN, Belda W, de Carvalho CHC, Laurenti MD, Passero LFD. New Immunological Markers in Chromoblastomycosis-The Importance of PD-1 and PD-L1 Molecules in Human Infection. J Fungi (Basel) 2023; 9:1172. [PMID: 38132773 PMCID: PMC10744586 DOI: 10.3390/jof9121172] [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: 10/24/2023] [Revised: 11/22/2023] [Accepted: 12/05/2023] [Indexed: 12/23/2023] Open
Abstract
The pathogenesis of chromoblastomycosis (CBM) is associated with Th2 and/or T regulatory immune responses, while resistance is associated with a Th1 response. However, even in the presence of IFN-γ, fungi persist in the lesions, and the reason for this persistence is unknown. To clarify the factors associated with pathogenesis, this study aimed to determine the polarization of the cellular immune response and the densities of cells that express markers of exhaustion in the skin of CBM patients. In the skin of patients with CBM, a moderate inflammatory infiltrate was observed, characterized primarily by the occurrence of histiocytes. Analysis of fungal density allowed us to divide patients into groups that exhibited low and high fungal densities; however, the intensity of the inflammatory response was not related to mycotic loads. Furthermore, patients with CBM exhibited a significant increase in the number of CD4+ and CD8+ cells associated with a high density of IL-10-, IL-17-, and IFN-γ-producing cells, indicating the presence of a chronic and mixed cellular immune response, which was also independent of fungal load. A significant increase in the number of PD-1+ and PD-L1+ cells was observed, which may be associated with the maintenance of the fungus in the skin and the progression of the disease.
Collapse
Affiliation(s)
- Italo N. Cavallone
- Institute of Biosciences, São Paulo State University (UNESP), Praça Infante Dom Henrique, s/n, São Vicente 11330-900, Brazil;
- Laboratory of Pathology of Infectious Diseases (LIM50), Department of Pathology, Medical School, São Paulo University, São Paulo 01246-903, Brazil
| | - Walter Belda
- Dermatology Department, Medical School, São Paulo University, Clinics Hospital, São Paulo 05403-000, Brazil; (W.B.J.); (C.H.C.d.C.); (M.D.L.)
| | - Caroline Heleno C. de Carvalho
- Dermatology Department, Medical School, São Paulo University, Clinics Hospital, São Paulo 05403-000, Brazil; (W.B.J.); (C.H.C.d.C.); (M.D.L.)
| | - Marcia D. Laurenti
- Dermatology Department, Medical School, São Paulo University, Clinics Hospital, São Paulo 05403-000, Brazil; (W.B.J.); (C.H.C.d.C.); (M.D.L.)
| | - Luiz Felipe D. Passero
- Institute of Biosciences, São Paulo State University (UNESP), Praça Infante Dom Henrique, s/n, São Vicente 11330-900, Brazil;
- Institute for Advanced Studies of Ocean (IEAMAR), São Paulo State University (UNESP), Rua João Francisco Bensdorp, 1178, São Vicente 11350-011, Brazil
| |
Collapse
|
35
|
Zhao H, Huang M, Jiang L. Potential Roles and Future Perspectives of Chitinase 3-like 1 in Macrophage Polarization and the Development of Diseases. Int J Mol Sci 2023; 24:16149. [PMID: 38003338 PMCID: PMC10671302 DOI: 10.3390/ijms242216149] [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: 10/16/2023] [Revised: 11/03/2023] [Accepted: 11/05/2023] [Indexed: 11/26/2023] Open
Abstract
Chitinase-3-like protein 1 (CHI3L1), a chitinase-like protein family member, is a secreted glycoprotein that mediates macrophage polarization, inflammation, apoptosis, angiogenesis, and carcinogenesis. Abnormal CHI3L1 expression has been associated with multiple metabolic and neurological disorders, including diabetes, atherosclerosis, and Alzheimer's disease. Aberrant CHI3L1 expression is also reportedly associated with tumor migration and metastasis, as well as contributions to immune escape, playing important roles in tumor progression. However, the physiological and pathophysiological roles of CHI3L1 in the development of metabolic and neurodegenerative diseases and cancer remain unclear. Understanding the polarization relationship between CHI3L1 and macrophages is crucial for disease progression. Recent research has uncovered the complex mechanisms of CHI3L1 in different diseases, highlighting its close association with macrophage functional polarization. In this article, we review recent findings regarding the various disease types and summarize the relationship between macrophages and CHI3L1. Furthermore, this article also provides a brief overview of the various mechanisms and inhibitors employed to inhibit CHI3L1 and disrupt its interaction with receptors. These endeavors highlight the pivotal roles of CHI3L1 and suggest therapeutic approaches targeting CHI3L1 in the development of metabolic diseases, neurodegenerative diseases, and cancers.
Collapse
Affiliation(s)
| | - Mingdong Huang
- College of Chemistry, Fuzhou University, Fuzhou 350116, China;
| | - Longguang Jiang
- College of Chemistry, Fuzhou University, Fuzhou 350116, China;
| |
Collapse
|
36
|
Gu J, Zhang X, Peng Z, Peng Z, Liao Z. A novel immune-related gene signature for predicting immunotherapy outcomes and survival in clear cell renal cell carcinoma. Sci Rep 2023; 13:18922. [PMID: 37919459 PMCID: PMC10622518 DOI: 10.1038/s41598-023-45966-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 10/26/2023] [Indexed: 11/04/2023] Open
Abstract
Clear cell renal carcinoma (ccRCC) is one of the most common cancers worldwide. In this study, a new model of immune-related genes was developed to predict the overall survival and immunotherapy efficacy in patients with ccRCC. Immune-related genes were obtained from the ImmPort database. Clinical data and transcriptomics of ccRCC samples were downloaded from GSE29609 and The Cancer Genome Atlas. An immune-related gene-based prognostic model (IRGPM) was developed using the least absolute shrinkage and selection operator regression algorithm and multivariate Cox regression. The reliability of the developed models was evaluated by Kaplan-Meier survival curves and time-dependent receiver operating characteristic curves. Furthermore, we constructed a nomogram based on the IRGPM and multiple clinicopathological factors, along with a calibration curve to examine the predictive power of the nomogram. Overall, this study investigated the association of IRGPM with immunotherapeutic efficacy, immune checkpoints, and immune cell infiltration. Eleven IRGs based on 528 ccRCC samples significantly associated with survival were used to construct the IRGPM. Remarkably, the IRGPM, which consists of 11 hub genes (SAA1, IL4, PLAUR, PLXNB3, ANGPTL3, AMH, KLRC2, NR3C2, KL, CSF2, and SEMA3G), was found to predict the survival of ccRCC patients accurately. The calibration curve revealed that the nomogram developed with the IRGPM showed high predictive performance for the survival probability of ccRCC patients. Moreover, the IRGPM subgroups showed different levels of immune checkpoints and immune cell infiltration in patients with ccRCC. IRGPM might be a promising biomarker of immunotherapeutic responses in patients with ccRCC. Overall, the established IRGPM was valuable for predicting survival, reflecting the immunotherapy response and immune microenvironment in patients with ccRCC.
Collapse
Affiliation(s)
- Jie Gu
- Department of Geriatric Urology, Xiangya International Medical Center, Xiangya Hospital, Central South University, Hunan Province, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China
| | - Xiaobo Zhang
- Department of Geriatric Urology, Xiangya International Medical Center, Xiangya Hospital, Central South University, Hunan Province, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China
| | - ZhangZhe Peng
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China
| | - Zhuoming Peng
- Department of Respiratory and Intensive Care Medicine, Union Shenzhen Hospital, Huazhong University of Science and Technology, Shenzhen, 518000, Guangdong Province, China
| | - Zhouning Liao
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China.
| |
Collapse
|
37
|
Lyu F, Wang L, Jia Y, Wang Y, Qi H, Dai Z, Zhou X, Zhu H, Li B, Xu Y, Liu J. Analysis of Zinc and Stromal Immunity in Disuse Osteoporosis: Mendelian Randomization and Transcriptomic Analysis. Orthop Surg 2023; 15:2947-2959. [PMID: 37752822 PMCID: PMC10622276 DOI: 10.1111/os.13840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 06/22/2023] [Accepted: 06/30/2023] [Indexed: 09/28/2023] Open
Abstract
OBJECTIVE Disuse osteoporosis is known to be primarily caused by a lack of exercise. However, the causal relationships between zinc and immunity and disuse osteoporosis remain unknown. This study investigated these relationships and their potential mechanisms. METHODS This study was an integrative study combining genome-wide association studies and transcriptomics. Two-sample Mendelian randomization analysis (MR) was used to analyze the causal relationships between exposures (zinc, immunity, physical activity) and the outcome (osteoporosis) with the aid of single-nucleotide polymorphisms (SNPs) as instrumental variables (IVs). Four models, MR-Egger, inverse variance weighted, weighted median and MR-Pleiotrophy RESidual Sum and Outlier (MRPRESSO), were used to calculate odds ratio values. Sensitivity and heterogeneity analyses were also performed using MRPRESSO and MR-Egger methods. The mRNA transcriptomic analysis was subsequently conducted. Zinc metabolism scores were acquired through single-sample Gene Set Enrichment Analysis algorithms. Stromal scores were obtained using the R Package "estimate" algorithms. Important Kyoto Encyclopedia of Genes and Genomes and Gene Ontology pathways were also derived through gene set variation analysis. Cytoscape software helped construct the transcription factor (TF)-mRNA-microRNA (miRNA) network. Virtual screening and molecular docking were performed. Polymerase chain reaction validation was also carried out in vivo. RESULTS Causal relationships were demonstrated between zinc and exercise (95% confidence interval [CI] = 1.30-2.95, p = 0.001), exercise and immunity (95% CI = 0.36-0.80, p = 0.002), exercise and osteoporosis (95% CI = 0.97-0.99, p = 0.0007), and immunity disorder and osteoporosis (95% CI = 1.30-2.03, p = 0.00002). One hundred and seventy-nine mRNAs in important modules were screened. Combining the differential expressional genes (DEGs) and the Boruta selection, six DEGs were screened (AHNAK, CSF2, ADAMTS12, SRA1, RUNX2, and SLC39A14). TF HOXC10 and miRNA hsa-miR-204 were predicted. Then, the TF-mRNA-miRNA network was successfully constructed. RUNX2 and SLC39A14 were identified as hub mRNAs in the TF-mRNA-miRNA network. Eventually, the novel small drug C6O4NH5 was designed according to the pharmacophore structure of SLC39A14. The docking energy for the novel drug was -5.83 kcal/mol. SLC39A14 and RUNX2 were downregulated (of statistical significance p-value < 0.05) in our animal experiment. CONCLUSION This study revealed that zinc had a protective causal relationship with disuse osteoporosis by promoting exercise and immunity. SLC39A14 and RUNX2 mRNA participated in this zinc-related mechanism.
Collapse
Affiliation(s)
- Fei Lyu
- College of OrthopedicsTianjin Medical UniversityTianjinChina
- Department of Joint SurgeryTianjin HospitalTianjinChina
- Orthopedic Center (Sports Medicine Center)Inner Mongolia People's HospitalHohhotChina
| | - Li Wang
- College of OrthopedicsTianjin Medical UniversityTianjinChina
- Department of Joint SurgeryTianjin HospitalTianjinChina
| | - Yiming Jia
- College of OrthopedicsTianjin Medical UniversityTianjinChina
- Department of Joint SurgeryTianjin HospitalTianjinChina
- Department of OrthopedicsChifeng Municipal HospitalChifengChina
| | - Yuanlin Wang
- Department of Joint SurgeryTianjin HospitalTianjinChina
- Tianjin Institute of AnesthesiologyTianjin Medical UniversityTianjinChina
| | - Haolan Qi
- School of MedicineNankai UniversityTianjinChina
| | - Zhengxu Dai
- College of OrthopedicsTianjin Medical UniversityTianjinChina
- Department of Joint SurgeryTianjin HospitalTianjinChina
| | - Xuyang Zhou
- College of OrthopedicsTianjin Medical UniversityTianjinChina
- Department of Joint SurgeryTianjin HospitalTianjinChina
| | - Haoran Zhu
- School of MedicineXi'an Jiaotong UniversityXianChina
| | - Bing Li
- College of OrthopedicsTianjin Medical UniversityTianjinChina
- Department of Joint SurgeryTianjin HospitalTianjinChina
| | - Yujing Xu
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of PharmacyTianjin Medical UniversityTianjinChina
| | - Jun Liu
- College of OrthopedicsTianjin Medical UniversityTianjinChina
- Department of Joint SurgeryTianjin HospitalTianjinChina
| |
Collapse
|
38
|
Zhang P, Wang Y, Miao Q, Chen Y. The therapeutic potential of PD-1/PD-L1 pathway on immune-related diseases: Based on the innate and adaptive immune components. Biomed Pharmacother 2023; 167:115569. [PMID: 37769390 DOI: 10.1016/j.biopha.2023.115569] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/14/2023] [Accepted: 09/18/2023] [Indexed: 09/30/2023] Open
Abstract
Currently, immunotherapy targeting programmed cell death 1 (PD-1) or programmed death ligand 1 (PD-L1) has revolutionized the treatment strategy of human cancer patients. Meanwhile, PD-1/PD-L1 pathway has also been implicated in the pathogenesis of many immune-related diseases, such as autoimmune diseases, chronic infection diseases and adverse pregnancy outcomes, by regulating components of the innate and adaptive immune systems. Given the power of the new therapy, a better understanding of the regulatory effects of PD-1/PD-L1 pathway on innate and adaptive immune responses in immune-related diseases will facilitate the discovery of novel biomarkers and therapeutic drug targets. Targeting this pathway may successfully halt or potentially even reverse these pathological processes. In this review, we discuss recent major advances in PD-1/PD-L1 axis regulating innate and adaptive immune components in immune-related diseases. We reveal that the impact of PD-1/PD-L1 axis on the immune system is complex and manifold and multi-strategies on the targeted PD-1/PD-L1 axis are taken in the treatment of immune-related diseases. Consequently, targeting PD-1/PD-L1 pathway, alone or in combination with other treatments, may represent a novel strategy for future therapeutic intervention on immune-related diseases.
Collapse
Affiliation(s)
- Peng Zhang
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention (China Medical University), Ministry of Education, Shenyang 110122, Liaoning, China; Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang 110122, Liaoning, China
| | - Yuting Wang
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention (China Medical University), Ministry of Education, Shenyang 110122, Liaoning, China; Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang 110122, Liaoning, China
| | - Qianru Miao
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention (China Medical University), Ministry of Education, Shenyang 110122, Liaoning, China; Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang 110122, Liaoning, China
| | - Ying Chen
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention (China Medical University), Ministry of Education, Shenyang 110122, Liaoning, China; Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang 110122, Liaoning, China.
| |
Collapse
|
39
|
Kitamura T. Tumour-associated macrophages as a potential target to improve natural killer cell-based immunotherapies. Essays Biochem 2023; 67:1003-1014. [PMID: 37313600 PMCID: PMC10539946 DOI: 10.1042/ebc20230002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 05/31/2023] [Accepted: 06/01/2023] [Indexed: 06/15/2023]
Abstract
Adoptive transfer of natural killer (NK) cells has been proposed as a novel immunotherapy for malignant tumours resistant to current therapeutic modalities. Several clinical studies have demonstrated that the NK cell-infusion is well tolerated without severe side effects and shows promising results in haematological malignancies. However, patients with malignant solid tumours do not show significant responses to this therapy. Such disappointing results largely arise from the inefficient delivery of infused NK cells and the impairment of their functions in the tumour microenvironment (TME). Tumour-associated macrophages (TAMs) are the most abundant stromal cells in the TME of most solid tumours, and a high TAM density correlates with poor prognosis of cancer patients. Although our knowledge of the interactions between TAMs and NK cells is limited, many studies have indicated that TAMs suppress NK cell cytotoxicity against cancer cells. Therefore, blockade of TAM functions can be an attractive strategy to improve NK cell-based immunotherapies. On the other hand, macrophages are reported to activate NK cells under certain circumstances. This essay presents our current knowledge about mechanisms by which macrophages regulate NK cell functions and discusses possible therapeutic approaches to block macrophage-mediated NK cell suppression.
Collapse
Affiliation(s)
- Takanori Kitamura
- MRC Centre for Reproductive Health, The University of Edinburgh, Edinburgh, EH16 4TJ, United Kingdom
| |
Collapse
|
40
|
Fernando V, Zheng X, Sharma V, Furuta S. Reprogramming of breast tumor-associated macrophages with modulation of arginine metabolism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.22.554238. [PMID: 37662241 PMCID: PMC10473631 DOI: 10.1101/2023.08.22.554238] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
HER2+ breast tumors have abundant immune-suppressive cells, including M2-type tumor associated macrophages (TAMs). While TAMs consist of the immune-stimulatory M1-type and immune-suppressive M2-type, M1/M2-TAM ratio is reduced in immune-suppressive tumors, contributing to their immunotherapy refractoriness. M1 vs. M2-TAM formation depends on differential arginine metabolism, where M1-TAMs convert arginine to nitric oxide (NO) and M2-TAMs convert arginine to polyamines (PAs). We hypothesize that such distinct arginine metabolism in M1- vs M2-TAMs is attributed to different availability of BH4 (NO synthase cofactor) and that its replenishment would reprogram M2-TAMs to M1-TAMs. Recently, we reported that sepiapterin (SEP), the endogenous BH4 precursor, elevates the expression of M1-TAM markers within HER2+ tumors. Here, we show that SEP restores BH4 levels in M2-TAMs, which then redirects arginine metabolism to NO synthesis and converts M2-TAMs to M1-TAMs. The reprogrammed TAMs exhibit full-fledged capabilities of antigen presentation and induction of effector T cells to trigger immunogenic cell death of HER2+ cancer cells. This study substantiates the utility of SEP in metabolic shift of HER2+ breast tumor microenvironment as a novel immunotherapeutic strategy.
Collapse
Affiliation(s)
- Veani Fernando
- Department of Cell & Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus, 3000 Arlington Ave. Toledo, OH 43614, USA
- Division of Rheumatology, University of Colorado, Anschutz Medical Campus Barbara Davis Center, Mail Stop B115, 1775 Aurora Court, Aurora, Colorado 80045
| | - Xunzhen Zheng
- Department of Cell & Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus, 3000 Arlington Ave. Toledo, OH 43614, USA
| | - Vandana Sharma
- Department of Cell & Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus, 3000 Arlington Ave. Toledo, OH 43614, USA
| | - Saori Furuta
- Department of Cell & Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus, 3000 Arlington Ave. Toledo, OH 43614, USA
- MetroHealth Medical Center, Case Western Reserve University School of Medicine, Case Comprehensive Cancer Center, 2500 MetroHealth Drive, Cleveland, OH 44109
| |
Collapse
|
41
|
Wang Y, Barrett A, Hu Q. Targeting Macrophages for Tumor Therapy. AAPS J 2023; 25:80. [PMID: 37589825 DOI: 10.1208/s12248-023-00845-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 07/27/2023] [Indexed: 08/18/2023] Open
Abstract
Macrophages, as one of the most abundant tumor-infiltrating cells, play an important role in tumor development and metastasis. The frequency and polarization of tumor-associated macrophages (TAMs) correlate with disease progression, tumor metastasis, and resistance to various treatments. Pro-inflammatory M1 macrophages hold the potential to engulf tumor cells. In contrast, anti-inflammatory M2 macrophages, which are predominantly present in tumors, potentiate tumor progression and immune escape. Targeting macrophages to modulate the tumor immune microenvironment can ameliorate the tumor-associated immunosuppression and elicit an anti-tumor immune response. Strategies to repolarize TAMs, deplete TAMs, and block inhibitory signaling hold great potential in tumor therapy. Besides, biomimetic carriers based on macrophages have been extensively explored to prolong circulation, enhance tumor-targeted delivery, and reduce the immunogenicity of therapeutics to augment therapeutic efficacy. Moreover, the genetic engineering of macrophages with chimeric antigen receptor (CAR) allows them to recognize tumor antigens and perform tumor cell-specific phagocytosis. These strategies will expand the toolkit for treating tumors, especially for solid tumors, drug-resistant tumors, and metastatic tumors. Herein, we introduce the role of macrophages in tumor progression, summarize the recent advances in macrophage-centered anticancer therapy, and discuss their challenges as well as future applications. Graphical abstract.
Collapse
Affiliation(s)
- Yixin Wang
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, WI, I 53705, Madison, U.S.A
- Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, WI, I 53705, Madison, U.S.A
- Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, WI, I 53705, Madison, U.S.A
| | - Allie Barrett
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, WI, I 53705, Madison, U.S.A
| | - Quanyin Hu
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, WI, I 53705, Madison, U.S.A..
- Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, WI, I 53705, Madison, U.S.A..
- Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, WI, I 53705, Madison, U.S.A..
| |
Collapse
|
42
|
Ma H, Zhang Z, Hu Q, Chen H, Wu G, Zhou Y, Xue Q. Shedding light on macrophage immunotherapy in lung cancer. J Cancer Res Clin Oncol 2023; 149:8143-8152. [PMID: 37052632 DOI: 10.1007/s00432-023-04740-z] [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: 02/13/2023] [Accepted: 04/02/2023] [Indexed: 04/14/2023]
Abstract
The search for therapeutic options for lung cancer continues to advance, with rapid advances in the search for therapies to improve patient prognosis. At present, systemic chemotherapy, immune checkpoint inhibitor therapy, antiangiogenic therapy, and targeted therapy for driver gene positivity are available in the clinic. Common clinical treatments fail to achieve desired outcomes due to immunosuppression of the tumor microenvironment (TME). Tumor immune evasion is mediated by cytokines, chemokines, immune cells, and other cells such as vascular endothelial cells within the tumor immune microenvironment. Tumor-associated macrophages (TAMs) are important immune cells in the TME, inducing tumor angiogenesis, encouraging tumor cell proliferation and migration, and suppressing antitumor immune responses. Thus, TAM targeting becomes the key to lung cancer immunotherapy. This review focuses on macrophage phenotype, polarization mechanism, role in lung cancer, and advances in macrophage centric immunotherapies.
Collapse
Affiliation(s)
- Huiyun Ma
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China
| | - Zhouwei Zhang
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China
| | - Qin Hu
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China
| | - Hongyu Chen
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China
| | - Gujie Wu
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China
| | - Youlang Zhou
- Research Central of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China.
| | - Qun Xue
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China.
| |
Collapse
|
43
|
Wang L, Wang S, Chiou S, Tsai J, Chai C, Tseng L, Lee J, Lin M, Huang S, Hsu S. HCV Core Protein-ISX Axis Promotes Chronic Liver Disease Progression via Metabolic Remodeling and Immune Suppression. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300644. [PMID: 37316966 PMCID: PMC10427408 DOI: 10.1002/advs.202300644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 04/13/2023] [Indexed: 06/16/2023]
Abstract
Chronic hepatitis C virus (HCV) infection is an important public health issue. However, knowledge on how the virus remodels the metabolic and immune response toward hepatic pathologic environment is limited. The transcriptomic and multiple evidences reveal that the HCV core protein-intestine-specific homeobox (ISX) axis promotes a spectrum of metabolic, fibrogenic, and immune modulators (e.g., kynurenine, PD-L1, and B7-2), regulating HCV-infection relevant pathogenic phenotype in vitro and in vivo. In a transgenic mice model, the HCV core protein-ISX axis enhance metabolic disturbance (particularly lipid and glucose metabolism) and immune suppression, and finally, chronic liver fibrosis in a high-fat diet (HFD)-induced disease model. Mechanistically, cells with HCV JFH-1 replicons upregulate ISX and, consequently, the expressions of metabolic, fibrosis progenitor, and immune modulators via core protein-induced nuclear factor-κB signaling. Conversely, cells with specific ISX shRNAi inhibit HCV core protein-induced metabolic disturbance and immune suppression. Clinically, the HCV core level is significantly correlated with ISX, IDOs, PD-L1, and B7-2 levels in HCC patients with HCV infection. Therefore, it highlights the significance of HCV core protein-ISX axis as an important mechanism in the development of HCV-induced chronic liver disease and can be a specific therapeutic target clinically.
Collapse
Affiliation(s)
- Li‐Ting Wang
- Department of Life ScienceNational Taiwan Normal UniversityTaipei116059Taiwan
- Center of Applied GenomicsKaohsiung Medical UniversityKaohsiung80708Taiwan
| | - Shen‐Nien Wang
- Graduate Institute of MedicineCollege of MedicineKaohsiung Medical UniversityKaohsiung80708Taiwan
- Division of General and Digestive SurgeryDepartment of SurgeryKaohsiung Medical University HospitalKaohsiung80708Taiwan
- Department of SurgeryCollege of MedicineKaohsiung Medical University HospitalKaohsiung80708Taiwan
| | - Shyh‐Shin Chiou
- Center of Applied GenomicsKaohsiung Medical UniversityKaohsiung80708Taiwan
- Division of Pediatric Hematology and Oncology, Department of PediatricsKaohsiung Medical University HospitalKaohsiung80708Taiwan
- Graduate Institute of Clinical Medicine, College of MedicineKaohsiung Medical UniversityKaohsiung80708Taiwan
| | - Jhih‐Peng Tsai
- Center of Applied GenomicsKaohsiung Medical UniversityKaohsiung80708Taiwan
| | - Chee‐Yin Chai
- Department of PathologyKaohsiung Medical University HospitalKaohsiung80708Taiwan
| | - Li‐Wen Tseng
- Graduate Institute of MedicineCollege of MedicineKaohsiung Medical UniversityKaohsiung80708Taiwan
| | - Jin‐Ching Lee
- Department of BiotechnologyCollege of Life ScienceNational Sun Yat‐sen UniversityKaohsiung804201Taiwan
| | - Ming‐Hong Lin
- Department of Microbiology and ImmunologySchool of MedicineCollege of MedicineKaohsiung Medical UniversityKaohsiung City80708Taiwan
- Department of Medical ResearchKaohsiung Medical University HospitalKaohsiung Medical UniversityKaohsiung80708Taiwan
| | - Shau‐Ku Huang
- National Institute of Environmental Health SciencesNational Health Research InstitutesMiaoli County35053Taiwan
- Department of Respirology & AllergyThird Affiliated Hospital of Shenzhen UniversityShenzhen518020China
- Department of MedicineDivision of Allergy and Clinical ImmunologyJohns Hopkins University School of MedicineBaltimoreMD21287USA
| | - Shih‐Hsien Hsu
- Graduate Institute of MedicineCollege of MedicineKaohsiung Medical UniversityKaohsiung80708Taiwan
- Department of Medical ResearchKaohsiung Medical University HospitalKaohsiung Medical UniversityKaohsiung80708Taiwan
| |
Collapse
|
44
|
Moeller A, Kurzrock R, Botta GP, Adashek JJ, Patel H, Lee S, Pabla S, Nesline MK, Conroy J, Sicklick JK, Kato S. Challenges and prospects of CSF1R targeting for advanced malignancies. Am J Cancer Res 2023; 13:3257-3265. [PMID: 37560003 PMCID: PMC10408490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 06/28/2023] [Indexed: 08/11/2023] Open
Abstract
CSF1R expression modulates tumor-associated macrophages, making CSF1R blockade an appealing immune-modulating therapeutic target. We evaluated the correlation between CSF1R tumor RNA expression and outcome (pan-cancer setting). RNA expression was ranked as a percentile (0-100) using a standardized internal reference population (735 tumors; 35 histologies). Among 514 patients, there was no difference in survival from biopsy between high and low CSF1R expressors (< 50 percentile versus ≥ 50 percentile rank). There was also no significant difference in median progression-free or overall survival (from treatment) based on CSF1R expression in 21 patients who received CSF1R inhibitors (all p values ≥ 0.08). Concurrent upregulation of ≥ 2 additional immune checkpoint markers (e.g. PD-L1, BTLA, CTLA4, LAG3, TIM3) was observed in all tumor samples with CSF1R expression ≥ 50th percentile. Pending further large prospective studies, patients with high tumor CSF1R expression may need treatment that co-targets the specific immune checkpoint pathways activated in order to impact outcome.
Collapse
Affiliation(s)
- Ann Moeller
- Sharp Rees-Stealy Medical GroupSan Diego, CA, USA
| | - Razelle Kurzrock
- Medical College of Wisconsin Cancer Center and Genomic Sciences and Precision Medicine CenterMilwaukee, WI, USA
- Worldwide Innovative Network (WIN) for Personalized Cancer TherapyParis, France
| | - Gregory P Botta
- Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer CenterLa Jolla, CA, USA
| | - Jacob J Adashek
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins HospitalBaltimore, MD, USA
| | - Hitendra Patel
- Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer CenterLa Jolla, CA, USA
| | - Suzanna Lee
- Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer CenterLa Jolla, CA, USA
| | | | | | | | - Jason K Sicklick
- Department of Surgery, Division of Surgical Oncology, UC San Diego School of MedicineSan Diego, CA, USA
| | - Shumei Kato
- Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer CenterLa Jolla, CA, USA
| |
Collapse
|
45
|
Song Z, Wang X, Liu X, Luo Y, Qiu J, Yin A, Liu Y, Yi H, Xiao Z, Li A. Targeting of Annexin A1 in Tumor-associated Macrophages as a therapeutic strategy for hepatocellular carcinoma. Biochem Pharmacol 2023; 213:115612. [PMID: 37209858 DOI: 10.1016/j.bcp.2023.115612] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 05/15/2023] [Accepted: 05/15/2023] [Indexed: 05/22/2023]
Abstract
Hepatocellular carcinoma (HCC) is a common aggressive, malignant tumor with limited treatment options. Currently, immunotherapies have low success rates in the treatment of HCC. Annexin A1 (ANXA1) is a protein related to inflammation, immunity and tumorigenesis. However, the role of ANXA1 in liver tumorigenesis remains unknown. Therefore, we sought to explore the feasibility of ANXA1 as a therapeutic target for HCC. Here, we analyzed ANXA1 expression and localization by HCC microarray and immunofluorescence experiments. Using an in vitro culture system, monocytic cell lines and primary macrophages were employed to investigate the biological functions of cocultured HCC cells and cocultured T cells. In vivo, Ac2-26, human recombinant ANXA1 (hrANXA1), and cell depletion (macrophages or CD8 + T cells) experiments were further conducted to investigate the role of ANXA1 in the tumor microenvironment (TME). We found that ANXA1 was overexpressed in mesenchymal cells, especially macrophages, in human liver cancer. Moreover, the expression of ANXA1 in mesenchymal cells was positively correlated with programmed death-ligand 1 expression. Knockdown of ANXA1 expression inhibited HCC cell proliferation and migration by increasing the M1/M2 macrophage ratio and promoting T-cell activation. hrANXA1 promoted malignant growth and metastasis in mice by increasing the infiltration and M2 polarization of tumor-associated macrophages (TAMs), generating an immunosuppressive TME and suppressing the antitumor CD8 + T-cell response. Together, our findings reveal that ANXA1 may be an independent prognostic factor for HCC and demonstrate the clinical translational significance of ANXA1 for tumor immunotherapy in HCC.
Collapse
Affiliation(s)
- Zhenghui Song
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, China; Research Center of Carcinogenesis and Targeted Therapy, Xiangya Hospital, Central South University, Changsha 410008, China; National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China; Department of Hepatology, Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510315, China
| | - Xue Wang
- Department of Hepatology, Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510315, China; School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Xinhui Liu
- Department of Hepatology, Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510315, China; School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Yue Luo
- Department of Hepatology, Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510315, China; School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Jieya Qiu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, China; Research Center of Carcinogenesis and Targeted Therapy, Xiangya Hospital, Central South University, Changsha 410008, China; National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Aiqi Yin
- Department of Obstetrics, Shenzhen Maternity and Child Healthcare Hospital, The First School of Clinical Medicine, Southern Medical University, Shenzhen, Guangdong 518028, China
| | - Yun Liu
- Department of Endocrinology and Metabolic Diseases, Affiliated Hospital (Clinical College) of Xiangnan University, Chenzhou 423000, China
| | - Hong Yi
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, China; Research Center of Carcinogenesis and Targeted Therapy, Xiangya Hospital, Central South University, Changsha 410008, China; National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Zhiqiang Xiao
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, China; Research Center of Carcinogenesis and Targeted Therapy, Xiangya Hospital, Central South University, Changsha 410008, China; National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China.
| | - Aimin Li
- Department of Hepatology, Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510315, China; School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China.
| |
Collapse
|
46
|
Zhang S, Xu Q, Sun W, Zhou J, Zhou J. Immunomodulatory effects of CDK4/6 inhibitors. Biochim Biophys Acta Rev Cancer 2023; 1878:188912. [PMID: 37182667 DOI: 10.1016/j.bbcan.2023.188912] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 04/23/2023] [Accepted: 05/10/2023] [Indexed: 05/16/2023]
Abstract
The dysregulation of the cell cycle is one of the hallmarks of cancer. Cyclin-dependent kinase 4 (CDK4) and CDK6 play crucial roles in regulating cell cycle and other cellular functions. CDK4/6 inhibitors have achieved great success in treating breast cancers and are currently being tested extensively in other tumor types as well. Accumulating evidence suggests that CDK4/6 inhibitors exert antitumor effects through immunomodulation aside from cell cycle arrest. Here we outline the immunomodulatory activities of CDK4/6 inhibitors, discuss the immune mechanisms of drug resistance and explore avenues to harness their immunotherapeutic potential when combined with immune checkpoint inhibitors (ICIs) or chimeric antigen receptor (CAR) T-cell therapy to improve the clinical outcomes.
Collapse
Affiliation(s)
- Shumeng Zhang
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qiaomai Xu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wenjia Sun
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jianya Zhou
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Jianying Zhou
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
47
|
Han J, Dong L, Wu M, Ma F. Dynamic polarization of tumor-associated macrophages and their interaction with intratumoral T cells in an inflamed tumor microenvironment: from mechanistic insights to therapeutic opportunities. Front Immunol 2023; 14:1160340. [PMID: 37251409 PMCID: PMC10219223 DOI: 10.3389/fimmu.2023.1160340] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 04/20/2023] [Indexed: 05/31/2023] Open
Abstract
Immunotherapy has brought a paradigm shift in the treatment of tumors in recent decades. However, a significant proportion of patients remain unresponsive, largely due to the immunosuppressive tumor microenvironment (TME). Tumor-associated macrophages (TAMs) play crucial roles in shaping the TME by exhibiting dual identities as both mediators and responders of inflammation. TAMs closely interact with intratumoral T cells, regulating their infiltration, activation, expansion, effector function, and exhaustion through multiple secretory and surface factors. Nevertheless, the heterogeneous and plastic nature of TAMs renders the targeting of any of these factors alone inadequate and poses significant challenges for mechanistic studies and clinical translation of corresponding therapies. In this review, we present a comprehensive summary of the mechanisms by which TAMs dynamically polarize to influence intratumoral T cells, with a focus on their interaction with other TME cells and metabolic competition. For each mechanism, we also discuss relevant therapeutic opportunities, including non-specific and targeted approaches in combination with checkpoint inhibitors and cellular therapies. Our ultimate goal is to develop macrophage-centered therapies that can fine-tune tumor inflammation and empower immunotherapy.
Collapse
Affiliation(s)
- Jiashu Han
- 4+4 Medical Doctor Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Dongcheng, Beijing, China
| | - Luochu Dong
- 4+4 Medical Doctor Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Dongcheng, Beijing, China
| | - Mengwei Wu
- Department of General Surgery, Peking Union Medical College Hospital (CAMS), Beijing, China
| | - Fei Ma
- Center for National Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
48
|
Wen J, Yin P, Su Y, Gao F, Wu Y, Zhang W, Chi P, Chen J, Zhang X. Knockdown of HMGB1 inhibits the crosstalk between oral squamous cell carcinoma cells and tumor-associated macrophages. Int Immunopharmacol 2023; 119:110259. [PMID: 37141670 DOI: 10.1016/j.intimp.2023.110259] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 01/30/2023] [Accepted: 04/28/2023] [Indexed: 05/06/2023]
Abstract
Tumor-associated macrophages (TAMs), the major component of the tumor microenvironment (TME), play distinctly different roles in different tumors. High mobility group box 1 (HMGB1), a nonhistone protein in the nucleus, can perform functions during inflammation and cancers. However, the role of HMGB1 in the crosstalk between oral squamous cell carcinoma (OSCC) cells and TAMs remains unclear. Here, we established a coculture system of TAMs and OSCC cells to explore the bidirectional effect and potential mechanism of HMGB1 in OSCC cell-TAM interactions. Our results showed that HMGB1 was significantly upregulated in OSCC tissues and positively associated with tumor progression, immune cell infiltration and macrophage polarization. Then, knocking down HMGB1 in OSCC cells inhibited the recruitment and polarization of cocultured TAMs. Moreover, the knockdown of HMGB1 in macrophages not only suppressed polarization, but also inhibited cocultured OSCC cell proliferation, migration and invasion in vitro and in vivo. Mechanistically, macrophages secreted higher levels of HMGB1 than OSCC cells, and dampening endogenous HMGB1 reduced HMGB1 secretion. Both OSCC cell-generated and macrophage-endogenous HMGB1 may regulate TAM polarization by promoting receptor TLR4 expression and NF-κB/p65 activation and enhancing IL-10/TGF-β expression. HMGB1 in OSCC cells may regulate macrophage recruitment via IL-6/STAT3. In addition, TAM-derived HMGB1 may affect aggressive phenotypes of cocultured OSCC cells by regulating the immunosuppressive microenvironment through the IL-6/STAT3/PD-L1 and IL-6/NF-κB/MMP-9 pathways. In conclusion, HMGB1 may regulate the crosstalk between OSCC cells and TAMs, including modulating macrophage polarization and attraction, enhancing cytokine secretion, and remodeling and creating an immunosuppressive TME to further affect OSCC progression.
Collapse
Affiliation(s)
- Jinlin Wen
- Beijing Institute of Dental Research, Beijing Stomatological Hospital & School of Stomatology, Capital Medical University, Beijing, 100050, China
| | - Panpan Yin
- Beijing Institute of Dental Research, Beijing Stomatological Hospital & School of Stomatology, Capital Medical University, Beijing, 100050, China
| | - Ying Su
- Beijing Institute of Dental Research, Beijing Stomatological Hospital & School of Stomatology, Capital Medical University, Beijing, 100050, China
| | - Feng Gao
- Beijing Institute of Dental Research, Beijing Stomatological Hospital & School of Stomatology, Capital Medical University, Beijing, 100050, China
| | - Yanlin Wu
- Beijing Institute of Dental Research, Beijing Stomatological Hospital & School of Stomatology, Capital Medical University, Beijing, 100050, China
| | - Wenbin Zhang
- Beijing Institute of Dental Research, Beijing Stomatological Hospital & School of Stomatology, Capital Medical University, Beijing, 100050, China
| | - Peng Chi
- Beijing Institute of Dental Research, Beijing Stomatological Hospital & School of Stomatology, Capital Medical University, Beijing, 100050, China
| | - Jiahui Chen
- Beijing Institute of Dental Research, Beijing Stomatological Hospital & School of Stomatology, Capital Medical University, Beijing, 100050, China
| | - Xinyan Zhang
- Beijing Institute of Dental Research, Beijing Stomatological Hospital & School of Stomatology, Capital Medical University, Beijing, 100050, China.
| |
Collapse
|
49
|
Zhuang S, Russell A, Guo Y, Xu Y, Xiao W. IFN-γ blockade after genetic inhibition of PD-1 aggravates skeletal muscle damage and impairs skeletal muscle regeneration. Cell Mol Biol Lett 2023; 28:27. [PMID: 37016287 PMCID: PMC10071770 DOI: 10.1186/s11658-023-00439-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 03/15/2023] [Indexed: 04/06/2023] Open
Abstract
BACKGROUND Innate immune responses play essential roles in skeletal muscle recovery after injury. Programmed cell death protein 1 (PD-1) contributes to skeletal muscle regeneration by promoting macrophage proinflammatory to anti-inflammatory phenotype transition. Interferon (IFN)-γ induces proinflammatory macrophages that appear to hinder myogenesis in vitro. Therefore, we tested the hypothesis that blocking IFN-γ in PD-1 knockout mice may dampen inflammation and promote skeletal muscle regeneration via regulating the macrophage phenotype and neutrophils. METHODS Anti-IFN-γ antibody was administered in PD-1 knockout mice, and cardiotoxin (CTX) injection was performed to induce acute skeletal muscle injury. Hematoxylin and eosin (HE) staining was used to view morphological changes of injured and regenerated skeletal muscle. Masson's trichrome staining was used to assess the degree of fibrosis. Gene expressions of proinflammatory and anti-inflammatory factors, fibrosis-related factors, and myogenic regulator factors were determined by real-time polymerase chain reaction (PCR). Changes in macrophage phenotype were examined by western blot and real-time PCR. Immunofluorescence was used to detect the accumulation of proinflammatory macrophages, anti-inflammatory macrophages, and neutrophils. RESULTS IFN-γ blockade in PD-1 knockout mice did not alleviate skeletal muscle damage or improve regeneration following acute cardiotoxin-induced injury. Instead, it exacerbated skeletal muscle inflammation and fibrosis, and impaired regeneration via inhibiting macrophage accumulation, blocking macrophage proinflammatory to anti-inflammatory transition, and enhancing infiltration of neutrophils. CONCLUSION IFN-γ is crucial for efficient skeletal muscle regeneration in the absence of PD-1.
Collapse
Affiliation(s)
- Shuzhao Zhuang
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
- Key Laboratory of Exercise and Health Sciences, Shanghai University of Sport, Ministry of Education, Shanghai, China
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Australia
| | - Aaron Russell
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Australia
| | - Yifan Guo
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
- Key Laboratory of Exercise and Health Sciences, Shanghai University of Sport, Ministry of Education, Shanghai, China
| | - Yingying Xu
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
- Key Laboratory of Exercise and Health Sciences, Shanghai University of Sport, Ministry of Education, Shanghai, China
| | - Weihua Xiao
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China.
- Key Laboratory of Exercise and Health Sciences, Shanghai University of Sport, Ministry of Education, Shanghai, China.
| |
Collapse
|
50
|
Elomaa H, Ahtiainen M, Väyrynen SA, Ogino S, Nowak JA, Lau MC, Helminen O, Wirta EV, Seppälä TT, Böhm J, Mecklin JP, Kuopio T, Väyrynen JP. Spatially resolved multimarker evaluation of CD274 (PD-L1)/PDCD1 (PD-1) immune checkpoint expression and macrophage polarisation in colorectal cancer. Br J Cancer 2023; 128:2104-2115. [DOI: 10.1038/s41416-023-02238-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 03/14/2023] [Accepted: 03/15/2023] [Indexed: 04/03/2023] Open
Abstract
Abstract
Background
The CD274 (PD-L1)/PDCD1 (PD-1) immune checkpoint interaction may promote cancer progression, but the expression patterns and prognostic significance of PD-L1 and PD-1 in the colorectal cancer microenvironment are inadequately characterised.
Methods
We used a custom 9-plex immunohistochemistry assay to quantify the expression patterns of PD-L1 and PD-1 in macrophages, T cells, and tumour cells in 910 colorectal cancer patients. We evaluated cancer-specific mortality according to immune cell subset densities using multivariable Cox regression models.
Results
Compared to PD-L1– macrophages, PD-L1+ macrophages were more likely M1-polarised than M2-polarised and located closer to tumour cells. PD-L1+ macrophage density in the invasive margin associated with longer cancer-specific survival [Ptrend = 0.0004, HR for the highest vs. lowest quartile, 0.52; 95% CI: 0.34–0.78]. T cell densities associated with longer cancer-specific survival regardless of PD-1 expression (Ptrend < 0.005 for both PD-1+ and PD-1– subsets). Higher densities of PD-1+ T cell/PD-L1+ macrophage clusters associated with longer cancer-specific survival (Ptrend < 0.005).
Conclusions
PD-L1+ macrophages show distinct polarisation profiles (more M1-like), spatial features (greater co-localisation with tumour cells and PD-1+ T cells), and associations with favourable clinical outcome. Our comprehensive multimarker assessment could enhance the understanding of immune checkpoints in the tumour microenvironment and promote the development of improved immunotherapies.
Collapse
|