1
|
Xu L, Qiu J, Ren Q, Wang D, Guo A, Wang L, Hou K, Wang R, Liu Y. Gold nanoparticles modulate macrophage polarization to promote skeletal muscle regeneration. Mater Today Bio 2025; 32:101653. [PMID: 40151803 PMCID: PMC11937682 DOI: 10.1016/j.mtbio.2025.101653] [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: 12/31/2024] [Revised: 02/20/2025] [Accepted: 03/09/2025] [Indexed: 03/29/2025] Open
Abstract
Skeletal muscle regeneration is a complex process that depends on the interplay between immune responses and muscle stem cell (MuSC) activity. Macrophages play a crucial role in this process, exhibiting distinct polarization states-M1 (pro-inflammatory) and M2 (anti-inflammatory)-that significantly affect tissue repair outcomes. Recent advancements in nanomedicine have positioned gold nanoparticles (Au NPs) as promising tools for modulating macrophage polarization and enhancing muscle regeneration. This review examines the role of Au NPs in influencing macrophage behavior, focusing on their physicochemical properties, biocompatibility, and mechanisms of action. We discuss how Au NPs can promote M2 polarization, facilitating tissue repair through modulation of cytokine production, interaction with cell surface receptors, and activation of intracellular signaling pathways. Additionally, we highlight the benefits of Au NPs on MuSC function, angiogenesis, and extracellular matrix remodeling. Despite the potential of Au NPs in skeletal muscle regeneration, challenges remain in optimizing nanoparticle design, developing targeted delivery systems, and understanding long-term effects. Future directions should focus on personalized medicine approaches and combination therapies to enhance therapeutic efficacy. Ultimately, this review emphasizes the transformative potential of Au NPs in regenerative medicine, offering hope for improved treatments for muscle injuries and diseases.
Collapse
Affiliation(s)
- Lining Xu
- JST Sarcopenia Research Centre, National Center for Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Jiahuang Qiu
- Research Center of Nano Technology and Application Engineering, School of Public Health,Dongguan Innovation Institute, Guangdong Medical University, Dongguan, 523808, China
| | - Quanzhong Ren
- JST Sarcopenia Research Centre, National Center for Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Dingding Wang
- JST Sarcopenia Research Centre, National Center for Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Anyi Guo
- JST Sarcopenia Research Centre, National Center for Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Ling Wang
- JST Sarcopenia Research Centre, National Center for Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
- Department of Radiology, National Center for Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Kedong Hou
- Department of Orthopedics, Beijing Pinggu District Hospital, Beijing, 101200, China
| | - Renxian Wang
- JST Sarcopenia Research Centre, National Center for Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Yajun Liu
- JST Sarcopenia Research Centre, National Center for Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
- Department of Spine Surgery, National Center for Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| |
Collapse
|
2
|
Yi YF, Fan ZQ, Liu C, Ding YT, Chen Y, Wen J, Jian XH, Li YF. Immunomodulatory effects and clinical application of exosomes derived from mesenchymal stem cells. World J Stem Cells 2025; 17:103560. [DOI: 10.4252/wjsc.v17.i3.103560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 01/16/2025] [Accepted: 02/17/2025] [Indexed: 03/21/2025] Open
Abstract
Exosomes (Exos) are extracellular vesicles secreted by cells and serve as crucial mediators of intercellular communication. They play a pivotal role in the pathogenesis and progression of various diseases and offer promising avenues for therapeutic interventions. Exos derived from mesenchymal stem cells (MSCs) have significant immunomodulatory properties. They effectively regulate immune responses by modulating both innate and adaptive immunity. These Exos can inhibit excessive inflammatory responses and promote tissue repair. Moreover, they participate in antigen presentation, which is essential for activating immune responses. The cargo of these Exos, including ligands, proteins, and microRNAs, can suppress T cell activity or enhance the population of immunosuppressive cells to dampen the immune response. By inhibiting lymphocyte proliferation, acting on macrophages, and increasing the population of regulatory T cells, these Exos contribute to maintaining immune and metabolic homeostasis. Furthermore, they can activate immune-related signaling pathways or serve as vehicles to deliver microRNAs and other bioactive substances to target tumor cells, which holds potential for immunotherapy applications. Given the immense therapeutic potential of MSC-derived Exos, this review comprehensively explores their mechanisms of immune regulation and therapeutic applications in areas such as infection control, tumor suppression, and autoimmune disease management. This article aims to provide valuable insights into the mechanisms behind the actions of MSC-derived Exos, offering theoretical references for their future clinical utilization as cell-free drug preparations.
Collapse
Affiliation(s)
- Yang-Fei Yi
- Department of Anatomy, Hunan Normal University School of Medicine, Changsha 410005, Hunan Province, China
| | - Zi-Qi Fan
- Department of Anatomy, Hunan Normal University School of Medicine, Changsha 410005, Hunan Province, China
| | - Can Liu
- Department of Anatomy, Hunan Normal University School of Medicine, Changsha 410005, Hunan Province, China
| | - Yi-Tong Ding
- Department of Anatomy, Hunan Normal University School of Medicine, Changsha 410005, Hunan Province, China
| | - Yao Chen
- Department of Anatomy, Hunan Normal University School of Medicine, Changsha 410005, Hunan Province, China
| | - Jie Wen
- Department of Anatomy, Hunan Normal University School of Medicine, Changsha 410005, Hunan Province, China
- Department of Pediatric Orthopedics, Hunan Provincial People’s Hospital, Changsha 410013, Hunan Province, China
| | - Xiao-Hong Jian
- Department of Anatomy, Hunan Normal University School of Medicine, Changsha 410005, Hunan Province, China
| | - Yu-Fei Li
- Department of Anatomy, Hunan Normal University School of Medicine, Changsha 410005, Hunan Province, China
| |
Collapse
|
3
|
Liu Z, Yang Y, Fang H, Cen B, Fan Y, Li J, Wang L, He S. Single-cell and spatial analyses reveal the effect of VSIG4 +S100A10 +TAMs on the immunosuppression of glioblastoma and anti-PD-1 immunotherapy. Int J Biol Macromol 2025:142415. [PMID: 40127797 DOI: 10.1016/j.ijbiomac.2025.142415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 03/04/2025] [Accepted: 03/21/2025] [Indexed: 03/26/2025]
Abstract
Therapeutic strategies aiming at the tumor immune microenvironment (TIME) hold promise for glioblastoma (GBM) treatment. However, adjuvant immunotherapies targeting checkpoint inhibitors just prove effective for a selected group of GBM patients. The extensive involvement of GBM-associated macrophages highlights their potential role in tumor behavior. In-depth exploration of the impact of macrophages on the efficacy of immunotherapy is crucial for enhancing treatment outcomes. In this study, we conducted a comprehensive analysis using bulk RNA-seq, single-cell RNA sequencing (scRNA-seq), and spatial transcriptomics to explore the heterogeneity of tumor-associated macrophages (TAMs) in GBM. Flow cytometry was employed to investigate the effects of VSIG4 on TAM phenotypes, and co-culture cellular assays were performed to evaluate its contribution to GBM malignancy. Integrating 16 patient samples, we examined the immunological significance of VSIG4+S100A10+TAMs. VSIG4 expression on macrophages is significantly upregulated and correlated with the TIME, promoting the polarization of macrophages towards M2 and facilitating GBM progression. Spatial transcriptomics and human samples multiplex immunofluorescence (mIF) confirmed the co-localization of VSIG4+S100A10+TAMs with various T cells, resulting in the inhibition of T cell immune responses and a reduction in anti-tumor immunity. Our findings demonstrate for the first time that VSIG4+S100A10+TAM is an independent prognostic indicator of poor outcome for GBM and markedly accumulates in patients exhibiting non-responsiveness to anti-PD-1 immunotherapy. Targeting this specific bifunctional subgroup can potentially open up new avenues for the immunotherapy of GBM.
Collapse
Affiliation(s)
- Ziyuan Liu
- Department of Pharmacy, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, Guangdong, China; National Medical Products Administration Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Yufan Yang
- National Medical Products Administration Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, Guangdong, China; Clinical Pharmacy Center, Nanfang Hospital, Southern Medical University, Guangzhou 510505, Guangdong, China
| | - Haiting Fang
- Department of Pharmacy, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, Guangdong, China; National Medical Products Administration Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Bohong Cen
- National Medical Products Administration Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, Guangdong, China; Clinical Pharmacy Center, Nanfang Hospital, Southern Medical University, Guangzhou 510505, Guangdong, China
| | - Yiqi Fan
- Department of Pharmacy, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, Guangdong, China; National Medical Products Administration Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Jianlong Li
- Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA; Department of Orthopedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China.
| | - Lijie Wang
- Department of Pharmacy, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, Guangdong, China.
| | - Shuai He
- Department of Pharmacy, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, Guangdong, China; National Medical Products Administration Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, Guangdong, China.
| |
Collapse
|
4
|
Liu M, Li TZ, Peng CM. The role of tumor-associated fibroblast-derived exosomes in chemotherapy resistance of colorectal cancer and its application prospect. Biochim Biophys Acta Gen Subj 2025:130796. [PMID: 40122307 DOI: 10.1016/j.bbagen.2025.130796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 03/03/2025] [Accepted: 03/18/2025] [Indexed: 03/25/2025]
Abstract
Colorectal cancer (CRC) is the second most common malignant tumor in the world. With its increasing incidence and younger age trend, its impact on human health has been paid more and more attention. Currently, we have a variety of chemotherapy drugs that can be used to treat colorectal cancer. However, the drug resistance of colorectal cancer has become a significant factor affecting its cure rate. Some studies have reported that exosomes are related to the occurrence of drug resistance. However, the exact mechanism is not precise. Therefore, we focused on the role of cancer associated-fibroblast-derived (CAFs-derived) exosomes in colorectal progression. It was found that cancer cells transmit information through exosome interaction and induce chemotherapy resistance by promoting epithelial-mesenchymal transition (EMT), up-regulating the Wnt/β-catenin signaling pathway, transforming growth factor-β1 (TGF-β1) pathway, promoting angiogenesis and other possible molecular mechanisms. In addition, in terms of clinical significance and therapeutic strategies, we explore the clinical relevance of CAFs-derived exosomes in colorectal cancer patients and their potential as potential biomarkers for predicting chemotherapy response. We also provide a new possible direction for overcoming chemotherapy resistance in colorectal cancer by targeting CAFs-derived exosomes.
Collapse
Affiliation(s)
- Meichen Liu
- The Second Clinical Medical College, Nanchang University, NanChang, China
| | - Teng-Zheng Li
- Department of Emergency, The second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, NanChang, China
| | - Cheng-Ming Peng
- Department of Emergency, The second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, NanChang, China.
| |
Collapse
|
5
|
Zhang L, Guo R, Chen M, Liu M, Liu Y, Yu Y, Zang J, Kong L, Li X. Inhibition of Ovarian Cancer Growth, Metastasis and Reverse the Tumor Microenvironment by Dual Drug-Loaded Polymer Micelle Targeting Tumor Microenvironment. Int J Nanomedicine 2025; 20:2969-2990. [PMID: 40098720 PMCID: PMC11911825 DOI: 10.2147/ijn.s507038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Accepted: 03/04/2025] [Indexed: 03/19/2025] Open
Abstract
Introduction Ovarian cancer is a malignant tumor that arises in the female reproductive system and is associated with a very high mortality rate. This is primarily due to the highly invasive nature of metastasis and recurrence. Transforming the immune environment from an immunosuppressive state to an anti-tumor state through the phenotypic transformation of tumor-associated macrophages is crucial for inhibiting the growth, metastasis, and recurrence of ovarian cancer. Methods A polymer micelle (RC-PH-Ms) containing paclitaxel (PTX) and honokiol (HNK) was designed based on high expression of reactive oxygen species in the tumor microenvironment. Once the micelles are actively targeted to the tumor microenvironment characterized by elevated levels of reactive oxygen species, the responsive bond is cleaved, thereby exposing the secondary targeting ligand C7R. The released PTX and HNK facilitate the transformation of relevant macrophages in the tumor microenvironment from an M2 phenotype to an M1 phenotype, which in turn inhibits tumor growth, invasion and metastasis, inhibit angiogenesis and reduce tumor recurrence. Results The effects of RC-PH-Ms on modulating the immune microenvironment and inhibiting tumor growth, invasion and metastasis, vascularization and recurrence were investigated both in vivo and in vitro. Conclusion RC-PH-Ms can significantly inhibit the metastasis and recurrence of ovarian cancer, which provides a new perspective for clinical treatment.
Collapse
Affiliation(s)
- Lu Zhang
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, People’s Republic of China
- Shenyang Key Laboratory of Targeted Delivery of Chinese Medicine, Liaoning University of Traditional Chinese Medicine, Shenyang, People’s Republic of China
| | - Ruibo Guo
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, People’s Republic of China
- Shenyang Key Laboratory of Targeted Delivery of Chinese Medicine, Liaoning University of Traditional Chinese Medicine, Shenyang, People’s Republic of China
| | - Muhan Chen
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, People’s Republic of China
- Shenyang Key Laboratory of Targeted Delivery of Chinese Medicine, Liaoning University of Traditional Chinese Medicine, Shenyang, People’s Republic of China
| | - Mo Liu
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, People’s Republic of China
- Shenyang Key Laboratory of Targeted Delivery of Chinese Medicine, Liaoning University of Traditional Chinese Medicine, Shenyang, People’s Republic of China
| | - Yang Liu
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, People’s Republic of China
- Shenyang Key Laboratory of Targeted Delivery of Chinese Medicine, Liaoning University of Traditional Chinese Medicine, Shenyang, People’s Republic of China
| | - Yang Yu
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, People’s Republic of China
- Shenyang Key Laboratory of Targeted Delivery of Chinese Medicine, Liaoning University of Traditional Chinese Medicine, Shenyang, People’s Republic of China
- Key Laboratory of Ministry of Education for TCM Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, People’s Republic of China
| | - Juan Zang
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, People’s Republic of China
- Shenyang Key Laboratory of Targeted Delivery of Chinese Medicine, Liaoning University of Traditional Chinese Medicine, Shenyang, People’s Republic of China
| | - Liang Kong
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, People’s Republic of China
- Shenyang Key Laboratory of Targeted Delivery of Chinese Medicine, Liaoning University of Traditional Chinese Medicine, Shenyang, People’s Republic of China
| | - Xuetao Li
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, People’s Republic of China
- Shenyang Key Laboratory of Targeted Delivery of Chinese Medicine, Liaoning University of Traditional Chinese Medicine, Shenyang, People’s Republic of China
| |
Collapse
|
6
|
Liu M, Wang Y, Zhang Y, Hu D, Tang L, Zhou B, Yang L. Landscape of small nucleic acid therapeutics: moving from the bench to the clinic as next-generation medicines. Signal Transduct Target Ther 2025; 10:73. [PMID: 40059188 PMCID: PMC11891339 DOI: 10.1038/s41392-024-02112-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 10/23/2024] [Accepted: 12/13/2024] [Indexed: 03/17/2025] Open
Abstract
The ability of small nucleic acids to modulate gene expression via a range of processes has been widely explored. Compared with conventional treatments, small nucleic acid therapeutics have the potential to achieve long-lasting or even curative effects via gene editing. As a result of recent technological advances, efficient small nucleic acid delivery for therapeutic and biomedical applications has been achieved, accelerating their clinical translation. Here, we review the increasing number of small nucleic acid therapeutic classes and the most common chemical modifications and delivery platforms. We also discuss the key advances in the design, development and therapeutic application of each delivery platform. Furthermore, this review presents comprehensive profiles of currently approved small nucleic acid drugs, including 11 antisense oligonucleotides (ASOs), 2 aptamers and 6 siRNA drugs, summarizing their modifications, disease-specific mechanisms of action and delivery strategies. Other candidates whose clinical trial status has been recorded and updated are also discussed. We also consider strategic issues such as important safety considerations, novel vectors and hurdles for translating academic breakthroughs to the clinic. Small nucleic acid therapeutics have produced favorable results in clinical trials and have the potential to address previously "undruggable" targets, suggesting that they could be useful for guiding the development of additional clinical candidates.
Collapse
Affiliation(s)
- Mohan Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yusi Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yibing Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Die Hu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lin Tang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Bailing Zhou
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Li Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
7
|
Zhao W, Wu Y, Wang Y, Li T, Liu Q, Hou Z. Exosomal miR-92a-3p modulates M2 macrophage polarization in colorectal cancer: implications for tumor migration and angiogenesis. Med Oncol 2025; 42:96. [PMID: 40059261 DOI: 10.1007/s12032-025-02635-2] [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: 12/06/2024] [Accepted: 02/13/2025] [Indexed: 03/29/2025]
Abstract
Colorectal cancer (CRC) is one of the most prevalent malignant neoplasms globally. Its development and metastasis are closely associated with the polarization of macrophages within the tumor microenvironment (TME). In particular, the polarization of M2-type macrophages has been demonstrated to be related to the promotion of tumor growth, migration, and angiogenesis. This study aims to investigate the role of miR-92a-3p in colon cancer-derived exosomes in regulating M2-type macrophage polarization by targeting EID2B and to elucidate the impact of this process on tumor migration and angiogenesis. MicroRNAs that were differentially expressed in plasma exosomes from CRC patients were initially identified through a search of the GEO database. The results were then verified by RT-qPCR using miR-92a-3p. The uptake of exosomes was observed via laser confocal microscopy, and the impact of miR-92a-3p on the polarization of exosomes and macrophages was examined through the use of RT-qPCR and WB. A bioinformatics analysis and a dual-luciferase reporter assay were employed to identify the downstream target of miR-92a-3p and to investigate its effect on the MAPK/ERK pathway. miR-92a-3p was upregulated in plasma exosomes of colon cancer patients and exhibited a positive correlation with lymph node metastasis. The results demonstrated that miR-92a-3p was capable of promoting M0 macrophage polarization toward the M2 phenotype, and of enhancing the migratory and invasive capacities of CRC cells, as well as their angiogenic potential in vitro. Bioinformatic analysis and experimental validation demonstrated that miR-92a-3p targeted EID2B and that this target gene was negatively correlated with M2-type macrophage polarization. The results demonstrated that miR-92a-3p promotes macrophage M2 polarization by activating the MAPK/ERK pathway. miR-92a-3p activates the MAPK/ERK pathway and induces macrophage M2 polarization by targeting EID2B, thereby promoting migration and angiogenesis in CRC. These findings offer new potential targets for the treatment of colon cancer.
Collapse
Affiliation(s)
- Wei Zhao
- School of Basic Medical Sciences, Chengde Medical University, Chengde, China
| | - Yudan Wu
- Affiliated Hospital of Chengde Medical University, Chengde, China
| | - Yixiao Wang
- School of Basic Medical Sciences, Chengde Medical University, Chengde, China
| | - Tongyi Li
- School of Basic Medical Sciences, Chengde Medical University, Chengde, China
| | - Qiuyan Liu
- Affiliated Hospital of Chengde Medical University, Chengde, China
| | - Zhiping Hou
- School of Basic Medical Sciences, Chengde Medical University, Chengde, China.
| |
Collapse
|
8
|
Zhang Y, Yan W, Wu L, Yu Z, Quan Y, Xie X. Different exosomes are loaded in hydrogels for the application in the field of tissue repair. Front Bioeng Biotechnol 2025; 13:1545636. [PMID: 40099037 PMCID: PMC11911322 DOI: 10.3389/fbioe.2025.1545636] [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: 12/15/2024] [Accepted: 02/12/2025] [Indexed: 03/19/2025] Open
Abstract
Exosomes are double-membrane vesicular nanoparticles in the category of extracellular vesicles, ranging in size from 30 to 150 nm, and are released from cells through a specific multi-step exocytosis process. Exosomes have emerged as promising tools for tissue repair due to their ability to transfer bioactive molecules that promote cell proliferation, differentiation, and tissue regeneration. However, the therapeutic application of exosomes is hindered by their rapid clearance from the body and limited retention at the injury site. To overcome these challenges, hydrogels, known for their high biocompatibility and porous structure, have been explored as carriers for exosomes. Hydrogels can provide a controlled release mechanism, prolonging the retention time of exosomes at targeted tissues, thus enhancing their therapeutic efficacy. This review focuses on the combination of different exosomes with hydrogels in the context of tissue repair. We first introduce the sources and functions of exosomes, particularly those from mesenchymal stem cells, and their roles in regenerative medicine. We then examine various types of hydrogels, highlighting their ability to load and release exosomes. Several strategies for encapsulating exosomes in hydrogels are discussed, including the impact of hydrogel composition and structure on exosome delivery efficiency. Finally, we review the applications of exosomes-loaded hydrogels in the repair of different tissues, such as skin, bone, cartilage, and nerve, and explore the challenges and future directions in this field. The combination of exosomes with hydrogels offers significant promise for advancing tissue repair strategies and regenerative therapies.
Collapse
Affiliation(s)
| | | | | | | | | | - Xin Xie
- College of Life Sciences, Northwest University, Xi’an, China
| |
Collapse
|
9
|
Zhang X, Zhao J, Qi G, Chen Y, Guo X, Zhang J, Chen S, Xu X, Feng J, Zhang Q, Gao B, Wang Z, Jin J. USP48 inhibits colorectal cancer progression and promotes M1-like macrophage polarization by stabilizing TAK1. Exp Cell Res 2025; 446:114469. [PMID: 39971179 DOI: 10.1016/j.yexcr.2025.114469] [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: 04/05/2024] [Revised: 02/16/2025] [Accepted: 02/16/2025] [Indexed: 02/21/2025]
Abstract
Ubiquitination and deubiquitination have emerged as pivotal regulators of the development of colorectal cancer (CRC). However, the precise role of USP48 in CRC tumorigenesis is poorly understood. In this study, immunohistochemistry, protein blotting, MTT assays, plate cloning, scratch assays, transwell assays, and Hoechst 33258 staining were utilized to assess the expression level of USP48 and its involvement in CRC. The interaction between USP48 and Transforming growth factor-β activated kinase-1(TAK1) was confirmed using co-IP. Additionally, the impact of deubiquitination on downstream signaling was determined through qRT-PCR. Furthermore, the associations between USP48 and tumor-associated macrophages within the tumor microenvironment were investigated using flow cytometry. The findings of our study demonstrated that USP48 expression is downregulated in CRC patients. Through deubiquitination, USP48 interacts with and stabilizes TAK1, leading to the inhibition of TAK1-triggered NF-κB activation and effectively suppresses CRC tumorigenesis. Moreover, this study showed a positive correlation between USP48 expression and M1-type TAM polarization, revealed the potential of USP48 as a molecular target for the effective treatment of CRC in the future.
Collapse
Affiliation(s)
- Xinwen Zhang
- Department of Gastrointestinal Surgery, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China; Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, Guangxi, China
| | - Jiawei Zhao
- Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, Guangxi, China; Guangxi Health Commission Key Laboratory of Tumor Immunology and Receptor-Targeted Drug Basic Research, Guilin Medical University, Guilin, Guangxi, China
| | - Guangying Qi
- Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, Guangxi, China
| | - Yujing Chen
- Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, Guangxi, China
| | - Xiaotong Guo
- Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, Guangxi, China
| | - Juzheng Zhang
- Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, Guangxi, China; Guangxi Health Commission Key Laboratory of Tumor Immunology and Receptor-Targeted Drug Basic Research, Guilin Medical University, Guilin, Guangxi, China
| | - Siqi Chen
- Department of Gastrointestinal Surgery, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China; Department of Oral Bioscience, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Xiaochen Xu
- Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, Guangxi, China
| | - Jiayuan Feng
- Department of Gastrointestinal Surgery, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
| | - Qinyu Zhang
- Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, Guangxi, China
| | - Bin Gao
- Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, Guangxi, China
| | - Zhenran Wang
- Department of Gastrointestinal Surgery, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China.
| | - Jiamin Jin
- Department of Gastrointestinal Surgery, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China; Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, Guangxi, China; Guangxi Health Commission Key Laboratory of Tumor Immunology and Receptor-Targeted Drug Basic Research, Guilin Medical University, Guilin, Guangxi, China.
| |
Collapse
|
10
|
Shi B, Du M, Chen Z. Advances in tumor immunotherapy targeting macrophages. Expert Rev Clin Immunol 2025; 21:259-276. [PMID: 39636579 DOI: 10.1080/1744666x.2024.2438721] [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: 04/08/2024] [Revised: 11/03/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024]
Abstract
INTRODUCTION In recent years, immunotherapy has shown significant therapeutic potential in patients with advanced tumors. However, only a small number of individuals benefit, mainly due to the tumor microenvironment (TME), which provides conditions for the development of tumors. Macrophages in TME, known as tumor-associated macrophages (TAM), are mainly divided into M1 anti-tumor and M2 pro-tumor phenotypes, which play a regulatory role in various stages of tumorigenesis, promote tumorigenesis and metastasis, and cause immunotherapy resistance. AREAS COVERED This review focuses on research strategies and preclinical/clinical research progress in translating TAM into antitumor phenotype by referring to the PubMed database for five years. These include small molecule chemotherapy drug development, metabolic regulation, gene editing, physical stimulation, nanotechnology-mediated combination therapy strategies, and chimeric antigen receptor-based immunotherapy. EXPERT OPINION It is necessary to explore the surface-specific receptors and cell signaling pathways of TAM further to improve the specificity and targeting of drugs and to strengthen research in the field of probes that can monitor changes in TAM in real time. In addition, the physical stimulation polarization strategy has the advantages of being noninvasive, economical, and stable and will have excellent clinical transformation value in the future.
Collapse
Affiliation(s)
- Binrui Shi
- Key Laboratory of Medical Imaging Precision Theranostics and Radiation Protection, College of Hunan Province, the Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China
- Institute of Medical Imaging, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Department of Medical imaging, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China
- The Seventh Affiliated Hospital, Hunan Veterans Administration Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China
| | - Meng Du
- Key Laboratory of Medical Imaging Precision Theranostics and Radiation Protection, College of Hunan Province, the Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China
- Institute of Medical Imaging, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The Seventh Affiliated Hospital, Hunan Veterans Administration Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China
| | - Zhiyi Chen
- Key Laboratory of Medical Imaging Precision Theranostics and Radiation Protection, College of Hunan Province, the Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China
- Institute of Medical Imaging, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
11
|
Chen J, Hu S, Liu J, Jiang H, Wang S, Yang Z. Exosomes: a double-edged sword in cancer immunotherapy. MedComm (Beijing) 2025; 6:e70095. [PMID: 39968497 PMCID: PMC11831209 DOI: 10.1002/mco2.70095] [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: 08/02/2024] [Revised: 01/08/2025] [Accepted: 01/10/2025] [Indexed: 02/20/2025] Open
Abstract
Over the past few decades, immunotherapy has emerged as a powerful strategy to overcome the limitations of conventional cancer treatments. The use of extracellular vesicles, particularly exosomes, which carry cargoes capable of modulating the immune response, has been extensively explored as a potential therapeutic approach in cancer immunotherapy. Exosomes can deliver their cargo to target cells, thereby influencing their phenotype and immunomodulatory functions. They exhibit either immunosuppressive or immune-activating characteristics, depending on their internal contents. These exosomes originate from diverse cell sources, and their internal contents can vary, suggesting that there may be a delicate balance between immune suppression and stimulation when utilizing them for immunotherapy. Therefore, a thorough understanding of the molecular mechanisms underlying the role of exosomes in cancer progression is essential. This review focuses on the molecular mechanisms driving exosome function and their impact on the tumor microenvironment (TME), highlighting the intricate balance between immune suppression and activation that must be navigated in exosome-based therapies. Additionally, it underscores the challenges and ongoing efforts to optimize exosome-based immunotherapies, thereby making a significant contribution to the advancement of cancer immunotherapy research.
Collapse
Affiliation(s)
- Jiayi Chen
- School of Life SciencesJilin UniversityChangchunChina
| | - Siyuan Hu
- School of Life SciencesJilin UniversityChangchunChina
| | - Jiayi Liu
- School of Life SciencesJilin UniversityChangchunChina
| | - Hao Jiang
- School of Life SciencesJilin UniversityChangchunChina
| | - Simiao Wang
- School of Life SciencesJilin UniversityChangchunChina
| | - Zhaogang Yang
- School of Life SciencesJilin UniversityChangchunChina
| |
Collapse
|
12
|
Yao Y, Li B, Chen C, Wang J, Yao F, Li Z. HVEM as a tumor-intrinsic regulator in non-small cell lung cancer: Suppression of metastasis via glycolysis inhibition and modulation of macrophage polarization. Pharmacol Res 2025; 213:107604. [PMID: 39832683 DOI: 10.1016/j.phrs.2025.107604] [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: 11/08/2024] [Revised: 01/12/2025] [Accepted: 01/14/2025] [Indexed: 01/22/2025]
Abstract
Herpes virus entry mediator (HVEM) is a novel costimulatory molecule which mediates stimulatory or inhibitory signals in immune responses which makes it an attractive target in cancer therapeutics. However, the role of tumor cell intrinsic HVEM on tumor biology remains largely unknown. In this study, We demonstrated that CK+HVEM+ tumor correlates with better survival using Multiplex immuno histochemistry (mIHC) in Human Lung Adenocarcinoma Tissue microarray. Next, we showed that HVEM knockdown promoted NSCLC cell invasion and metastasis in vitro whereas exhibited no effect on proliferation. Conversely, HVEM overexpression results in the opposite phenotype. Meanwhile, the conclusion were further confirmed in vivo experiment that overexpression of HVEM reduced the invasion and metastasis of NSCLC whereas no effect on tumor mass. Besides, vivo experiment showed that M1 TAMs in the HVEM overxrpression group was increased and the proportion of M2 macrophages was decreased compared to the vector group. Mechanistically, The C-terminal 228-283 amino acid segment of HVEM protein interacts with the N-terminal 1-383 amino acid segment of MPRIP protein, inhibiting its downstream glycolysis signaling pathway and suppressing NSCLC cells progression. In addition, macrophage coculture assay suggested that HVEM overexpression inhibited M2 macrophage polarization through GM-CSF/GM-CSFRα axis. In summary, our study has demonstrated that tumor cell intrinsic HVEM is a potential tumour metastasis suppressor, which may serve as a potential target for immunotherapy.
Collapse
MESH Headings
- Lung Neoplasms/pathology
- Lung Neoplasms/metabolism
- Lung Neoplasms/immunology
- Lung Neoplasms/genetics
- Glycolysis
- Receptors, Tumor Necrosis Factor, Member 14/metabolism
- Receptors, Tumor Necrosis Factor, Member 14/genetics
- Humans
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Non-Small-Cell Lung/metabolism
- Carcinoma, Non-Small-Cell Lung/immunology
- Animals
- Cell Line, Tumor
- Macrophages/metabolism
- Macrophages/immunology
- Female
- Mice
- Male
- Mice, Nude
- Mice, Inbred BALB C
Collapse
Affiliation(s)
- Yuanshan Yao
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Bin Li
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Chunji Chen
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Jing Wang
- Department of Thoracic Surgery, Huadong Hospital affiliated to Fudan University, Shanghai 200040, China
| | - Feng Yao
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; Shanghai Institute of Thoracic Oncology, Shanghai 200030, China.
| | - Zhigang Li
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; Shanghai Institute of Thoracic Oncology, Shanghai 200030, China.
| |
Collapse
|
13
|
Zhang Q, Zhang Y, Guo S, Wang H. Emerging insights into the role of microRNAs regulation of ferroptosis in hepatocellular carcinoma. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167642. [PMID: 39734007 DOI: 10.1016/j.bbadis.2024.167642] [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: 09/08/2024] [Revised: 12/03/2024] [Accepted: 12/23/2024] [Indexed: 12/31/2024]
Abstract
Hepatocellular carcinoma (HCC) is a major type of liver cancer and an important cause of cancer death. It has been reported that the hepatocyte death plays an important role in HCC. Ferroptosis is an iron-dependent programmed cell death characterized by the accumulation of free iron and lipid peroxidation. A series of studies have shown that ferroptosis contributes to the occurrence and development of HCC. MicroRNAs (miRNAs) are non-coding RNAs with a length of approximately 222 nt. In recent years, miRNAs have been shown to participate in regulating ferroptosis to play a vital role in HCC, but the related mechanisms are not fully understood. This review summarized the current understanding of ferroptosis, as well as the biogenesis and function of miRNAs, and focused on the role of miRNAs regulation of ferroptosis in HCC, with the hope of providing new targets and ideas for the treatment of HCC.
Collapse
Affiliation(s)
- Qi Zhang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Yingdan Zhang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Shiyun Guo
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Honggang Wang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China.
| |
Collapse
|
14
|
Dou X, Feng C, Li J, Jiang E, Shang Z. Extracellular vesicle-mediated crosstalk in tumor microenvironment dominates tumor fate. Trends Cell Biol 2025; 35:230-247. [PMID: 39327161 DOI: 10.1016/j.tcb.2024.08.008] [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: 03/15/2024] [Revised: 08/16/2024] [Accepted: 08/29/2024] [Indexed: 09/28/2024]
Abstract
The tumor microenvironment (TME) is a complex and heterogeneous system containing various cells cooperating and competing with each other. Extracellular vesicles (EVs) differing in form and content are important intercellular communication mediators in the TME. Previous studies have focused on the cargoes within EVs rather than on the donors from which they originate and the recipient cells that exert their effects. Therefore, we provide here a detailed overview of the important roles of EVs in shaping tumor fate, highlighting their various mechanisms of intercellular dialog within the TME. We evaluate recent advances and also raise unresolved challenges to provide new ideas for clinical treatment strategies using EVs.
Collapse
Affiliation(s)
- Xinyu Dou
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan 430079, China
| | - Chunyu Feng
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan 430079, China
| | - Ji Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan 430079, China
| | - Erhui Jiang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan 430079, China; Department of Oral and Maxillofacial Head Neck Surgery, School & Hospital of Stomatology, Wuhan University, 237 Luoyu Road, Hongshan District, Wuhan 430079, China.
| | - Zhengjun Shang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan 430079, China; Department of Oral and Maxillofacial Head Neck Surgery, School & Hospital of Stomatology, Wuhan University, 237 Luoyu Road, Hongshan District, Wuhan 430079, China.
| |
Collapse
|
15
|
Ge Y, Jiang L, Dong Q, Xu Y, Yam JWP, Zhong X. Exosome-mediated Crosstalk in the Tumor Immune Microenvironment: Critical Drivers of Hepatocellular Carcinoma Progression. J Clin Transl Hepatol 2025; 13:143-161. [PMID: 39917466 PMCID: PMC11797817 DOI: 10.14218/jcth.2024.00302] [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: 08/23/2024] [Revised: 11/05/2024] [Accepted: 11/08/2024] [Indexed: 02/09/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a significant global health issue, ranking as the sixth most prevalent malignancy and the fourth leading cause of cancer-related mortality worldwide. Despite advancements in therapeutic strategies, mortality rates for HCC remain high. The tumor immune microenvironment (TIME) plays a vital role in HCC progression by influencing tumor cell survival and growth. Recent studies highlight the essential role of exosomes in mediating intercellular communication within the TIME, particularly in interactions among tumor cells, immune cells, and fibroblasts. These interactions drive critical aspects of tumor development, including immune escape, angiogenesis, drug resistance, and metastasis. A detailed understanding of the molecular mechanisms by which exosomes modulate the TIME is essential for developing targeted therapies. This review systematically evaluated the roles and regulatory mechanisms of exosomes within the TIME of HCC, examining the impact of both HCC-derived and non-HCC-derived exosomes on various cellular components within the TIME. It emphasized their regulatory effects on cell phenotypes and functions, as well as their roles in HCC progression. The review also explored the potential applications of exosome-based immunotherapies, offering new insights into improving therapeutic strategies for HCC.
Collapse
Affiliation(s)
- Yifei Ge
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Lixue Jiang
- Department of Breast Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Qingfu Dong
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yi Xu
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, China
- Fujian Provincial Key Laboratory of Tumor Biotherapy, Fuzhou, Fujian, China
- Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian, China
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Judy Wai Ping Yam
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Xiangyu Zhong
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
16
|
Yang X, Fang R, Li X, Kong W, Jin Y, Jiao R, Liu Z, Zhang M, Peng Q, Zhang Y, Song N. Engineered Nanovesicles for the Precise and Noninvasive Treatment of Deep Osteomyelitis Caused by MRSA Infection with Enhanced Immune Response. ACS APPLIED MATERIALS & INTERFACES 2025; 17:11795-11810. [PMID: 39945439 DOI: 10.1021/acsami.4c20893] [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: 02/28/2025]
Abstract
The clinical treatment of hospital-acquired persistent osteomyelitis caused by methicillin-resistant Staphylococcus aureus (MRSA) presents two major challenges: ineffective drug delivery into deep tissues and counteracting the rapid establishment of an immunosuppressive microenvironment. Indeed, MRSA can evade immunosurveillance and undermine both innate and adaptive immune responses. Herein, the engineered nanovesicles, functioning by combining sonodynamic therapy (SDT) with immune modulation, were constructed for the precise and noninvasive removal of MRSA in deep tissue and activation of the antimicrobial immune response using a newly engineered nanovesicle. Macrophage-derived M1 phenotypic microvesicles (M1-MW) internalized vancomycin-cross-linked micelles with the acoustic sensitizer indocyanine green (ICG) (VCG micelles). The vesicles of M1-MW were grafted with PEGylated mannose, allowing for targeted accumulation at the infection site. The VCG micelles were responsive to the highly reducing environment and released ICG to generate ROS after exposure to ultrasounds. This effect was combined with the presence of vancomycin to kill MRSA. In an osteomyelitis infection model, we observed an improved survival rate and reprogramming of macrophages to a pro-inflammatory M1 phenotype. The latter promoted T-cell activation and immune defense against MRSA-camouflaged homologous cell-transferred infections. Thus, our study presents a noninvasive and efficient treatment (VCG@MMW) for deep osteomyelitis with improved bacterial clearance and reduced risk of recurrence with enhanced immune response.
Collapse
Affiliation(s)
- Xingyue Yang
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261000, P. R. China
| | - Ren Fang
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261000, P. R. China
| | - Xiaotian Li
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261000, P. R. China
| | - Weihao Kong
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261000, P. R. China
| | - Yubao Jin
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261000, P. R. China
| | - Ruohan Jiao
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261000, P. R. China
| | - Zhenggong Liu
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261000, P. R. China
| | - Meiqi Zhang
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261000, P. R. China
| | - Qixian Peng
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261000, P. R. China
| | - Yumiao Zhang
- School of Chemical Engineering and Technology, Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin 300350, P. R. China
| | - Ningning Song
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang 261000, P. R. China
| |
Collapse
|
17
|
Cui J, Lin S, Zhang M. Resveratrol loaded microglia-derived exosomes attenuate astrogliasis by restoring mitochondrial function to reduce spinal cord injury. Chem Biol Interact 2025; 408:111407. [PMID: 39884494 DOI: 10.1016/j.cbi.2025.111407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 01/23/2025] [Accepted: 01/28/2025] [Indexed: 02/01/2025]
Abstract
The activation of astrocytes in the injured lesion induces the progression of spinal cord injury (SCI). However, adverse side-effects during systemic administration have limited applications. Exosomes (Exos) are an emerging clinical treatment method that exerts anti-inflammatory effects by reducing pro-inflammatory factors and promoting functional recovery. Exosomes exhibit great potential as carriers of traditional Chinese medicine, attributed to their high delivery efficiency to internalized and targeted accumulation in inflammatory tissues. Herein, We synthesized resveratrol loaded microglia-derived exosomes (R-MDEs) for highly efficient accumulation and infiltration in the injured spinal cord. In vitro and in vivo experiments suggested that R-MDEs effectively accumulated in A1 astrocytes, inhibited reactive oxygen species (ROS) and glial scar formation by reprogramming the metabolic astrocytes. R-MDEs achieve a synergistic therapeutic effect of immunomodulation and neuroprotection, thereby shedding new light on the application of Exos and provides great potential for SCI.
Collapse
Affiliation(s)
- Jingwen Cui
- Department of Neurosurgery, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, PR China; Liaoning Provincial Collaborative Innovation Center for Medical Testing and Drug Research, Jinzhou Medical University, Jinzhou, PR China; Key Laboratory of Medical Tissue Engineering of Liaoning Province, Jinzhou Medical University, Jinzhou, PR China
| | - Sen Lin
- Liaoning Provincial Collaborative Innovation Center for Medical Testing and Drug Research, Jinzhou Medical University, Jinzhou, PR China; Key Laboratory of Medical Tissue Engineering of Liaoning Province, Jinzhou Medical University, Jinzhou, PR China.
| | - Mingyuan Zhang
- Department of Neurosurgery, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, PR China.
| |
Collapse
|
18
|
Yao Y, Li B, Wang J, Chen C, Gao W, Li C. A novel HVEM-Fc recombinant protein for lung cancer immunotherapy. J Exp Clin Cancer Res 2025; 44:62. [PMID: 39979981 PMCID: PMC11841141 DOI: 10.1186/s13046-025-03324-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 02/10/2025] [Indexed: 02/22/2025] Open
Abstract
BACKGROUND The ubiquitously expressed transmembrane protein, Herpesvirus Entry Mediator (HVEM), functions as a molecular switch, capable of both activating and inhibiting the immune response depending on its interacting ligands. HVEM-Fc is a novel recombinant fusion protein with the potential to eradicate tumor cells. METHODS The anti-tumor efficacy of HVEM-Fc was evaluated in C57BL/6 mice-bearing lung cancer models: a syngeneic model and an orthotopic model of mouse lung cancer. Additionally, patient-derived organoids were employed in conjunction with T cell co-culture systems. To investigate the underlying mechanisms, a comprehensive array of techniques was utilized, including single-cell RNA sequencing, spatial transcriptomics, bulk RNA sequencing, and flow cytometry. Furthermore, the anti-tumor effects of HVEM-Fc in combination with Programmed Death-1 (PD-1) inhibitors were assessed. Finally, mouse immune cell depletion antibodies were used to elucidate the underlying mechanisms of action. RESULTS In vivo, 1 mg/kg HVEM-Fc demonstrated effective inhibition of tumor growth and metastasis in C57BL/6 mice bearing lung cancer model and a KP orthotopic model of mouse lung cancer. Multi-omics analysis showed that HVEM-Fc induced an immune-stimulatory microenvironment. Notably, the combination of HVEM-Fc with a PD-1 inhibitor demonstrated the most potent inhibition of tumor cell growth. In vitro, HVEM-Fc was validated to eradicate tumor cells through the activation of T cells in both non-small cell lung cancer (NSCLC) organoids and T cell co-culture models. CONCLUSIONS Our data demonstrate that HVEM-Fc exerts a strong signal that augments and prolongs T-cell activity in both murine models and human NSCLC organoid models. Moreover, the combination of HVEM-Fc with a PD-1 inhibitor yields the most effective anti-tumor outcomes.
Collapse
Affiliation(s)
- Yuanshan Yao
- Department of Thoracic Surgery, Huadong Hospital Affiliated to Fudan University, Shanghai, 200041, China
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
- Shanghai Institute of Thoracic Oncology, Shanghai, 200030, China
| | - Bin Li
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
- Shanghai Institute of Thoracic Oncology, Shanghai, 200030, China
| | - Jing Wang
- Department of Thoracic Surgery, Huadong Hospital Affiliated to Fudan University, Shanghai, 200041, China
| | - Chunji Chen
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
- Shanghai Institute of Thoracic Oncology, Shanghai, 200030, China
| | - Wen Gao
- Department of Thoracic Surgery, Huadong Hospital Affiliated to Fudan University, Shanghai, 200041, China.
| | - Chunguang Li
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
- Shanghai Institute of Thoracic Oncology, Shanghai, 200030, China.
| |
Collapse
|
19
|
Kuhl GC, Tangney M. Bacterial-Mediated In Situ Engineering of Tumour-Associated Macrophages for Cancer Immunotherapy. Cancers (Basel) 2025; 17:723. [PMID: 40075571 PMCID: PMC11899205 DOI: 10.3390/cancers17050723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 01/29/2025] [Accepted: 02/12/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND/OBJECTIVES Tumour-associated macrophages (TAMs) are critical components of the tumour microenvironment (TME), significantly influencing cancer progression and treatment resistance. This review aims to explore the innovative use of engineered bacteria to reprogram TAMs, enhancing their anti-tumour functions and improving therapeutic outcomes. METHODS We conducted a systematic review following a predefined protocol. Multiple databases were searched to identify relevant studies on TAMs, their phenotypic plasticity, and the use of engineered bacteria for reprogramming. Inclusion and exclusion criteria were applied to select studies, and data were extracted using standardised forms. Data synthesis was performed to summarise the findings, focusing on the mechanisms and therapeutic benefits of using non-pathogenic bacteria to modify TAMs. RESULTS The review summarises the findings that engineered bacteria can selectively target TAMs, promoting a shift from the tumour-promoting M2 phenotype to the tumour-fighting M1 phenotype. This reprogramming enhances pro-inflammatory responses and anti-tumour activity within the TME. Evidence from various studies indicates significant tumour regression and improved immune responses following bacterial therapy. CONCLUSIONS Reprogramming TAMs using engineered bacteria presents a promising strategy for cancer therapy. This approach leverages the natural targeting abilities of bacteria to modify TAMs directly within the tumour, potentially improving patient outcomes and offering new insights into immune-based cancer treatments. Further research is needed to optimise these methods and assess their clinical applicability.
Collapse
Affiliation(s)
- Gabriela Christina Kuhl
- Cancer Research @UCC, College of Medicine and Health, University College Cork, T12 K8AF Cork, Ireland;
| | - Mark Tangney
- Cancer Research @UCC, College of Medicine and Health, University College Cork, T12 K8AF Cork, Ireland;
- APC Microbiome Ireland, University College Cork, T12 YT20 Cork, Ireland
| |
Collapse
|
20
|
Yuan Q, Jia L, Yang J, Li W. The role of macrophages in liver metastasis: mechanisms and therapeutic prospects. Front Immunol 2025; 16:1542197. [PMID: 40034694 PMCID: PMC11872939 DOI: 10.3389/fimmu.2025.1542197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 01/21/2025] [Indexed: 03/05/2025] Open
Abstract
Metastasis is a hallmark of advanced cancer, and the liver is a common site for secondary metastasis of many tumor cells, including colorectal, pancreatic, gastric, and prostate cancers. Macrophages in the tumor microenvironment (TME) promote tumor cell metastasis through various mechanisms, including angiogenesis and immunosuppression, and play a unique role in the development of liver metastasis. Macrophages are affected by a variety of factors. Under conditions of hypoxia and increased acidity in the TME, more factors are now found to promote the polarization of macrophages to the M2 type, including exosomes and amino acids. M2-type macrophages promote tumor cell angiogenesis through a variety of mechanisms, including the secretion of factors such as VEGF, IL-1β, and TGF-β1. M2-type macrophages are subjected to multiple regulatory mechanisms. They also interact with various cells within the tumor microenvironment to co-regulate certain conditions, including the creation of an immunosuppressive microenvironment. This interaction promotes tumor cell metastasis, drug resistance, and immune escape. Based on the advent of single-cell sequencing technology, further insights into macrophage subpopulations in the tumor microenvironment may help in exploring new therapeutic targets in the future. In this paper, we will focus on how macrophages affect the TME, how tumor cells and macrophages as well as other immune cells interact with each other, and further investigate the mechanisms involved in liver metastasis of tumor cells and their potential as therapeutic targets.
Collapse
Affiliation(s)
| | | | | | - Wei Li
- *Correspondence: Jiahua Yang, ; Wei Li,
| |
Collapse
|
21
|
Yang P, Li Y. Progress of KRAS G12C inhibitors in the treatment of refractory colorectal cancer and strategies for drug resistance response. Invest New Drugs 2025:10.1007/s10637-025-01514-x. [PMID: 39956882 DOI: 10.1007/s10637-025-01514-x] [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: 09/29/2024] [Accepted: 02/07/2025] [Indexed: 02/18/2025]
Abstract
Colorectal cancer is the third most prevalent cancer in the world. Early screening and detection of tumours, active surgical radical treatment, postoperative adjuvant chemotherapy, targeted therapy, and immunotherapy are performed based on pathological staging and immunohistochemistry. Even with these measures, the 5-year survival rate of colorectal cancer is only 65%, and a considerable number of patients still experience tumour recurrence or even metastasis. The KRAS G12C mutation accounts for 3 to 4% of refractory colorectal cancer (advanced or metastatic colorectal cancer), and it was once believed that KRAS did not have a drug target until the emergence of KRAS G12C inhibitors provided targeted treatment for KRAS-mutated colorectal cancer. However, KRAS G12C inhibitors only produce moderate efficacy, and resistance occurs after a short remission. The mechanism of drug resistance in tumour cells is complex and diverse, and existing research has limited understanding of it. This review aims to elucidate the clinical trial progress of KRAS G12C inhibitors in refractory colorectal cancer, the research progress of drug resistance mechanisms, and the combined treatment strategies for drug resistance.
Collapse
Affiliation(s)
- Peiyuan Yang
- Department of Gastrointestinal Surgery, China-Japan Union Hospital of Jilin University, Erdao District, 126 Xiantai Street, Changchun, 130000, Jilin, China
| | - Yongchao Li
- Department of Gastrointestinal Surgery, China-Japan Union Hospital of Jilin University, Erdao District, 126 Xiantai Street, Changchun, 130000, Jilin, China.
| |
Collapse
|
22
|
Meng Y, Yao Z, Ke X, Hu M, Ren H, Gao S, Zhang H. Extracellular vesicles-based vaccines: Emerging immunotherapies against cancer. J Control Release 2025; 378:438-459. [PMID: 39667569 DOI: 10.1016/j.jconrel.2024.12.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 12/03/2024] [Accepted: 12/05/2024] [Indexed: 12/14/2024]
Abstract
Cancer vaccines are promising therapeutic approaches to enhance specific T-cell immunity against most solid tumors. By stimulating anti-tumor immunity, clearing minimal residual disease, and minimizing adverse effects, these vaccines target tumor cells and are effective when combined with immune checkpoint blockade or other immunotherapies. However, the development of tumor cell-based vaccines faces quality issues due to poor immunogenicity, tumor heterogeneity, a suppressive tumor immune microenvironment, and ineffective delivery methods. In contrast, extracellular vesicles (EVs), naturally released by cells, are considered the ideal drug carriers and vaccine platforms. EVs offer highly organ-specific targeting, induce broader and more effective immune responses, and demonstrate superior tissue delivery ability. The development of EV vaccines is crucial for advancing cancer immunotherapy. Compared to cell-based vaccines, EV vaccines produced under Good Manufacturing Practices (GMP) offer advantages such as high safety, ease of preservation and transport, and a wide range of sources. This review summarizes the latest research findings on EV vaccine and potential applications in this field. It also highlights novel neoantigens for the development of EV vaccines against cancer.
Collapse
Affiliation(s)
- Yuhua Meng
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China; State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, and Institute of Precision Cancer Medicine and Pathology, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Zhimeng Yao
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, and Institute of Precision Cancer Medicine and Pathology, School of Medicine, Jinan University, Guangzhou, Guangdong, China; Department of Urology Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Xiurong Ke
- Department of Surgery, Laboratory for Translational Surgical Oncology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Mengyuan Hu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, and Institute of Precision Cancer Medicine and Pathology, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Hongzheng Ren
- Gongli Hospital of Shanghai Pudong New Area, Department of Pathology, Shanghai, China
| | - Shegan Gao
- College of Clinical Medicine, The First Affiliated Hospital of Henan University of Science and Technology, Henan Key Laboratory of Cancer Epigenetics, Luoyang, Henan, China.
| | - Hao Zhang
- Gongli Hospital of Shanghai Pudong New Area, Department of Pathology, Shanghai, China; Department of Pathology, and Institute of Precision Cancer Medicine and Pathology, School of Medicine, State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou, Guangdong, China; Department of Thoracic Surgery and General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China.
| |
Collapse
|
23
|
Gao Y, Liu J, Wu M, Zhang Y, Wang M, Lyu Q, Zhang W, Zhou Y, Cheuk YC, Wang X, Liu Y, Wang W, Tu W. Photosensitive Hybrid γδ-T Exosomes for Targeted Cancer Photoimmunotherapy. ACS NANO 2025; 19:4251-4268. [PMID: 39862206 PMCID: PMC11803918 DOI: 10.1021/acsnano.4c11024] [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: 08/12/2024] [Revised: 01/14/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025]
Abstract
Melanoma is the most aggressive type of skin cancers. Traditional chemotherapy and radiotherapy have limited effectiveness and can lead to systemic side effects. Photodynamic therapy (PDT) is a photoresponsive cancer therapy based on photosensitizers to generate reactive oxygen species (ROS) to eradicate tumor cells. Our previous study showed that exosomes derived from human γδ-T cells (γδ-T exosomes) could control Epstein-Barr virus-associated tumors. Here, we combined γδ-T exosomes and PDT for targeted photoimmunotherapy by membrane fusion of γδ-T exosomes and Chlorin e6 (Ce6)-loaded liposomes. The functional surface proteins, such as CCR5 and PD-1, on the hybrid exosomes mediated the specific binding of hybrid exosomes toward melanoma tissues. The cytolytic molecules, such as granzyme A, granzyme B, perforin, and granulysin from γδ-T exosomes, induced specific apoptosis of cancer cells without harming normal cells. In response to light irradiation, ROS generation inside melanoma cells synergized with cytolytic molecules to induce apoptosis and promote immunogenic cancer cell death (ICD). The subsequently released damage-associated molecular patterns (DAMPs) could stimulate human dendritic cell maturation and induce melanoma antigen-specific CD4+ and CD8+ T-cell responses, thereby enhancing antitumor immunity. This study provides a promising strategy by combining γδ-T exosomes and PDT for photoimmunotherapy, thereby expanding the clinical applications of γδ-T exosome therapy for cancer patients.
Collapse
Affiliation(s)
- Yifan Gao
- Department
of Paediatrics & Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Jinzhao Liu
- State
Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China
- Department
of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Dr.
Li Dak-Sum Research Centre, The University
of Hong Kong, Hong Kong SAR, China
| | - Meicen Wu
- State
Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China
- Department
of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Dr.
Li Dak-Sum Research Centre, The University
of Hong Kong, Hong Kong SAR, China
| | - Yanmei Zhang
- Department
of Paediatrics & Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Manni Wang
- Department
of Paediatrics & Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Qingyang Lyu
- State
Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China
- Department
of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Dr.
Li Dak-Sum Research Centre, The University
of Hong Kong, Hong Kong SAR, China
| | - Wenyue Zhang
- Department
of Paediatrics & Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Yang Zhou
- State
Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China
- Department
of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Dr.
Li Dak-Sum Research Centre, The University
of Hong Kong, Hong Kong SAR, China
| | - Yin Celeste Cheuk
- Department
of Paediatrics & Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Xiwei Wang
- Department
of Paediatrics & Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Yinping Liu
- Department
of Paediatrics & Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Weiping Wang
- State
Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China
- Department
of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Dr.
Li Dak-Sum Research Centre, The University
of Hong Kong, Hong Kong SAR, China
| | - Wenwei Tu
- Department
of Paediatrics & Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
24
|
Li J, Zhao Y, Wu X, Zou Y, Liu Y, Ma H. Choline kinase alpha regulates autophagy-associated exosome release to promote glioma cell progression. Biochem Biophys Res Commun 2025; 746:151269. [PMID: 39778250 DOI: 10.1016/j.bbrc.2024.151269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/30/2024] [Accepted: 12/30/2024] [Indexed: 01/11/2025]
Abstract
Glioma is the most common primary intracranial malignant tumor in adults, with a poor prognosis. Exosomes released by tumor cells play a crucial role in tumor development, metastasis, angiogenesis, and other biological processes. Despite this significance, the precise molecular mechanisms governing exosome secretion and their impact on tumor progression remain incompletely understood. While Choline Kinase Alpha (CHKA) has been implicated in promoting various types of tumors, its specific role in glioma pathogenesis remains unclear. Our study initially demonstrates that CHKA enhances the proliferation, migration, and invasion abilities of glioma cells. Interestingly, CHKA also stimulates the release of exosomes from glioma cells. Mechanistically, reduced CHKA expression hampers exosome secretion by elevating autophagy levels in gliomas, whereas counteracting the autophagy elevation resulting from CHKA downregulation restores the release of exosomes. Notably, exosomes derived from glioma cells with normal CHKA expression exhibit a greater capacity to promote glioma progression compared to those derived from cells with low CHKA expression. Overall, our findings suggest that CHKA modulates exosome secretion via an autophagy-dependent pathway, thereby facilitating the proliferation, migration, and invasion of glioma cells.
Collapse
Affiliation(s)
- Jialin Li
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
| | - Yang Zhao
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
| | - Xiao Wu
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
| | - Yourui Zou
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Yang Liu
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Hui Ma
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, China.
| |
Collapse
|
25
|
Yang Y, Peng Y, Li Y, Shi T, Xu N, Luan Y, Yin C. Sestrin2 balances mitophagy and apoptosis through the PINK1-Parkin pathway to attenuate severe acute pancreatitis. Cell Signal 2025; 126:111518. [PMID: 39577789 DOI: 10.1016/j.cellsig.2024.111518] [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: 06/27/2024] [Revised: 09/08/2024] [Accepted: 11/18/2024] [Indexed: 11/24/2024]
Abstract
Mitophagy serves as a mitochondrial quality control mechanism to maintain the homeostasis of mitochondria and the intracellular environment. Studies have shown that there is a close relationship between mitophagy and apoptosis. Sestrin2 (Sesn2) is a highly conserved class of stress-inducible proteins that play important roles in reducing oxidative stress damage, inflammation, and apoptosis. However, the potential mechanism of how Sesn2 regulates mitophagy and apoptosis in severe acute pancreatitis (SAP) remains unclear. In the study, RAW264.7 (macrophage cell Line) cellular inflammation model established by lipopolysaccharide (LPS) treatment as well as LPS and CAE-induced SAP mouse model (wild-type and Sen2 Knockout mouse) were used. Our study showed that LPS stimulation significantly increased the level of Sesn2 in RAW264.7 cells, Sesn2 increased mitochondrial membrane potential, decreased inflammation levels, mitochondrial superoxide levels and apoptosis, and also promoted monocyte macrophages toward the M2 anti-inflammatory phenotype, suggesting a protective effect of Sesn2 on mitochondria. Further, Sesn2 increased mitophagy and decreased apoptosis via modulating the PINK1-Parkin signaling. Meanwhile, knockout of Sesn2 exacerbated pancreatic, mitochondrial damage and inflammation in a mouse model of SAP. In addition, the protective effect of Sesn2 against SAP was shown to be associated with mitophagy conducted by the PINK1-Parkin pathway via inhibiting apoptosis. These findings reveal that Sesn2 in balancing mitochondrial autophagy and apoptosis by modulating the PINK1-Parkin signaling may present a new therapeutic strategy for the treatment of SAP.
Collapse
Affiliation(s)
- Yuxi Yang
- Department of Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100026, China
| | - Yiqiu Peng
- Department of Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100026, China
| | - Yingying Li
- Department of Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100026, China
| | - Tingjuan Shi
- Department of Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100026, China
| | - Ning Xu
- Department of Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100026, China
| | - Yingyi Luan
- Department of Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100026, China.
| | - Chenghong Yin
- Department of Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100026, China.
| |
Collapse
|
26
|
Song Y, Lei L, Cai X, Wei H, Yu CY. Immunomodulatory Peptides for Tumor Treatment. Adv Healthc Mater 2025; 14:e2400512. [PMID: 38657003 DOI: 10.1002/adhm.202400512] [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: 02/08/2024] [Revised: 04/07/2024] [Indexed: 04/26/2024]
Abstract
Peptides exhibit various biological activities, including biorecognition, cell targeting, and tumor penetration, and can stimulate immune cells to elicit immune responses for tumor immunotherapy. Peptide self-assemblies and peptide-functionalized nanocarriers can reduce the effect of various biological barriers and the degradation by peptidases, enhancing the efficiency of peptide delivery and improving antitumor immune responses. To date, the design and development of peptides with various functionalities have been extensively reviewed for enhanced chemotherapy; however, peptide-mediated tumor immunotherapy using peptides acting on different immune cells, to the knowledge, has not yet been summarized. Thus, this work provides a review of this emerging subject of research, focusing on immunomodulatory anticancer peptides. This review introduces the role of peptides in the immunomodulation of innate and adaptive immune cells, followed by a link between peptides in the innate and adaptive immune systems. The peptides are discussed in detail, following a classification according to their effects on different innate and adaptive immune cells, as well as immune checkpoints. Subsequently, two delivery strategies for peptides as drugs are presented: peptide self-assemblies and peptide-functionalized nanocarriers. The concluding remarks regarding the challenges and potential solutions of peptides for tumor immunotherapy are presented.
Collapse
Affiliation(s)
- Yang Song
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Longtianyang Lei
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Xingyu Cai
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Hua Wei
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Cui-Yun Yu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
- Affiliated Hospital of Hunan Academy of Chinese Medicine, Hunan Academy of Chinese Medicine, Changsha, 410013, China
| |
Collapse
|
27
|
Sun X, Chen X, Ni Y, Li X, Song J, Wang J, Xu S, Shu W, Chen M. Latexin (LXN) enhances tumor immune surveillance in mice by inhibiting Treg cells through the macrophage exosome pathway. Int J Biol Macromol 2025; 289:138822. [PMID: 39694381 DOI: 10.1016/j.ijbiomac.2024.138822] [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: 09/10/2024] [Revised: 12/12/2024] [Accepted: 12/14/2024] [Indexed: 12/20/2024]
Abstract
Latexin (LXN) is a secreted protein with a molecular weight of 29 KD, which is considered a tumor suppressor and plays an important role in the inflammatory immune response. LXN is highly expressed in macrophages and regulates macrophage polarity and tumor immune escape, demonstrating excellent clinical potential. However, its mechanism is still unclear. In this study, a macrophage-T cell co-culture system is established to clarify the secretion of macrophage LXN into the extracellular through exosomes. The results indicate that LXN in macrophage-derived exosomes is functional, that is, LXN-enriched exosome inhibits CD4+T cell differentiation into Treg cells in vitro and in vivo, and exhibits good tumor suppressive effects. Based on this discovery, a biomimetic nanoparticle loaded with LXN protein (MØ@LXN-NPS) is designed and synthesized. Furthermore, the MØ@LXN-NPS shows excellent performance in both in vivo and in vitro, especially in enhancing tumor immune surveillance by inhibiting Treg cells in tumor microenvironment, thus exhibiting excellent anti-tumor activity. This study provides a demonstration for the transition of biomolecules from functional research to engineering applications. The excellent performance of MØ@LXN-NPS in tumor immune regulation suggests that the engineered biomimetic nanomedicine has good clinical application prospects.
Collapse
Affiliation(s)
- Xuchen Sun
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, China
| | - Xuanming Chen
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, China
| | - Yuanting Ni
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, China
| | - Xiuzhen Li
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, China
| | - Jiaqi Song
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, China
| | - Jingzhu Wang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, China
| | - Shaohua Xu
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, China
| | - Wei Shu
- College of Intelligent Medicine and Biotechnology, Guilin Medical University, Guilin, China
| | - Ming Chen
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, China.
| |
Collapse
|
28
|
Jia W, Wu Y, Xie Y, Yu M, Chen Y. Advanced Polymeric Nanoparticles for Cancer Immunotherapy: Materials Engineering, Immunotherapeutic Mechanism and Clinical Translation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2413603. [PMID: 39797474 DOI: 10.1002/adma.202413603] [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: 09/10/2024] [Revised: 12/13/2024] [Indexed: 01/13/2025]
Abstract
Cancer immunotherapy, which leverages immune system components to treat malignancies, has emerged as a cornerstone of contemporary therapeutic strategies. Yet, critical concerns about the efficacy and safety of cancer immunotherapies remain formidable. Nanotechnology, especially polymeric nanoparticles (PNPs), offers unparalleled flexibility in manipulation-from the chemical composition and physical properties to the precision control of nanoassemblies. PNPs provide an optimal platform to amplify the potency and minimize systematic toxicity in a broad spectrum of immunotherapeutic modalities. In this comprehensive review, the basics of polymer chemistry, and state-of-the-art designs of PNPs from a physicochemical standpoint for cancer immunotherapy, encompassing therapeutic cancer vaccines, in situ vaccination, adoptive T-cell therapies, tumor-infiltrating immune cell-targeted therapies, therapeutic antibodies, and cytokine therapies are delineated. Each immunotherapy necessitates distinctively tailored design strategies in polymeric nanoplatforms. The extensive applications of PNPs, and investigation of their mechanisms of action for enhanced efficacy are particularly focused on. The safety profiles of PNPs and clinical research progress are discussed. Additionally, forthcoming developments and emergent trends of polymeric nano-immunotherapeutics poised to transform cancer treatment paradigms into clinics are explored.
Collapse
Affiliation(s)
- Wencong Jia
- School of Medicine, Shanghai University, Shanghai China, 200444, China
| | - Ye Wu
- School of Medicine, Shanghai University, Shanghai China, 200444, China
| | - Yujie Xie
- School of Medicine, Shanghai University, Shanghai China, 200444, China
| | - Meihua Yu
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, China
- Shanghai Institute of Materdicine, Shanghai, 200051, China
| |
Collapse
|
29
|
Pan Y, Yin Q, Wang Z, Wu G, Liu K, Li X, Liu J, Zeng J, Lin B, Li W, Zhu M, Li M. AFP shields hepatocellular carcinoma from macrophage phagocytosis by regulating HuR-mediated CD47 translocation in cellular membrane. Transl Oncol 2025; 52:102240. [PMID: 39667226 PMCID: PMC11699289 DOI: 10.1016/j.tranon.2024.102240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 09/27/2024] [Accepted: 12/07/2024] [Indexed: 12/14/2024] Open
Abstract
OBJECTIVES Alpha fetoprotein(AFP) overexpression connecting with macrophage dysfunction remain poorly defined. In this study, explore AFP regulates macrophage immunomodulation in hepatocellular carcinoma(HCC) through comprehensive in vitro and in vivo studies. METHODS Immunohistochemical and immunofluorescence staining was used to analyze the relativity of AFP and cellular membrane CD47 expression in clinical 30 HCC tissues, and the expression of AFP and CD47 in HCC cells. The intelligent living-cell high-throughput imaging analyzer was applied to dynamically track and image of macrophages to phagocytize HCC cells. The effect of AFP on regulating the level of CD47 in cellular membrane and growth of tumor in vivo was performed by animal experiment. The association of AFP and CD47 in HCC cells was detected by single cell analysis. RESULTS The present results indicated that AFP upregulated the localization of CD47 on the HCC cell surface. CD47 overexpression stimulates HCC to escape immune surveillance by transmitting "don't eat me" signals to macrophages, lead to inhibit macrophage to phagocytize HCC cells. Mechanistically, the results demonstrated that AFP enhanced CD47 membrane translocation by interacting with Hu-Antigen R(HuR), an RNA-binding protein that regulates mRNA stability and translation. AFP alters the subcellular distribution of HuR, increasing its cytoplasmic accumulation and binding to CD47 transcript. CONCLUSIONS AFP enhanced CD47 membrane translocation by interacting with HuR. These findings proved that AFP could inhibit macrophage to phagocytize HCC cells by upregulating the localization of CD47 on the HCC cell surface. Combination of AFP with CD47 blockade may be a potential therapeutic strategy for HCC treatment.
Collapse
Affiliation(s)
- Yinglian Pan
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical University, Haikou, Hainan Province, PR China; Department of Medical Oncology, the First Affiliated Hospital of Hainan Medical University, Haikou, Hainan Province, PR China
| | - Qiushi Yin
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical University, Haikou, Hainan Province, PR China
| | - Zhaoliang Wang
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical University, Haikou, Hainan Province, PR China; Department of Laboratory, Affiliated Hainan Hospital of Hainan Medical University (Hainan General Hospital), Haikou, PR China
| | - Gang Wu
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical University, Haikou, Hainan Province, PR China; Department of Laboratory, Affiliated Hainan Hospital of Hainan Medical University (Hainan General Hospital), Haikou, PR China
| | - Kun Liu
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical University, Haikou, Hainan Province, PR China
| | - Xiaowei Li
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical University, Haikou, Hainan Province, PR China
| | - Jinchen Liu
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical University, Haikou, Hainan Province, PR China
| | - Jiangzheng Zeng
- Department of Medical Oncology, the First Affiliated Hospital of Hainan Medical University, Haikou, Hainan Province, PR China
| | - Bo Lin
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical University, Haikou, Hainan Province, PR China
| | - Wei Li
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical University, Haikou, Hainan Province, PR China
| | - Mingyue Zhu
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical University, Haikou, Hainan Province, PR China.
| | - Mengsen Li
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical University, Haikou, Hainan Province, PR China; Department of Medical Oncology, the Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan Province, PR China.
| |
Collapse
|
30
|
Abedi A, Moosazadeh Moghaddam M, Kachuei R, Imani Fooladi AA. Exosomes as a Therapeutic Strategy in Cancer: Potential Roles as Drug Carriers and Immune Modulators. Biochim Biophys Acta Rev Cancer 2025; 1880:189238. [PMID: 39674417 DOI: 10.1016/j.bbcan.2024.189238] [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: 07/22/2024] [Revised: 12/04/2024] [Accepted: 12/05/2024] [Indexed: 12/16/2024]
Abstract
Exosome-based cancer immunotherapy is advancing quickly on the concept of artificially activating the immune system to combat cancer. They can mechanistically change the tumor microenvironment, increase immune responses, and function as efficient drug delivery vehicles because of their inherent bioactivity, low toxicity, and immunogenicity. Accurate identification of the mechanisms of action of exosomes in tumor environments, along with optimization of their isolation, purification, and characterization methods, is necessary to increase clinical applications. Exosomes can be modified through cargo loading and surface modification to enhance their therapeutic applications, either before or after the donor cells' isolation. These engineered exosomes can directly target tumor cells at the tumor site or indirectly activate innate and adaptive immune responses in the tumor microenvironment. This approach is particularly effective when combined with traditional cancer immunotherapy techniques such as vaccines, immune checkpoints, and CAR-T cells. It can improve anti-tumor responses, induce long-term immunity, and address the limitations of traditional therapies, such as poor penetration in solid tumors and immunosuppressive environments. This review aims to provide a comprehensive and detailed overview of the direct role of engineered exosomes as drug delivery systems and their immunomodulatory effects on tumors as an indirect approach to fighting cancer. Additionally, it will discuss novel immunotherapy options.
Collapse
Affiliation(s)
- Azam Abedi
- Tissue Engineering and Regenerative Medicine Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mehrdad Moosazadeh Moghaddam
- Tissue Engineering and Regenerative Medicine Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Reza Kachuei
- Molecular Biology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Abbas Ali Imani Fooladi
- Applied Microbiology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
31
|
Gangadaran P, Onkar A, Rajendran RL, Goenka A, Oh JM, Khan F, Nagarajan AK, Muthu S, Krishnan A, Hong CM, Ahn BC. Noninvasive in vivo imaging of macrophages: understanding tumor microenvironments and delivery of therapeutics. Biomark Res 2025; 13:20. [PMID: 39865337 PMCID: PMC11770947 DOI: 10.1186/s40364-025-00735-9] [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: 10/22/2024] [Accepted: 01/21/2025] [Indexed: 01/28/2025] Open
Abstract
Macrophages are pivotal in the body's defense and response to inflammation. They are present in significant numbers and are widely implicated in various diseases, including cancer. While molecular and histological techniques have advanced our understanding of macrophage biology, their precise function within the cancerous microenvironments remains underexplored. Enhancing our knowledge of macrophages and the dynamics of their extracellular vesicles (EVs) in cancer development can potentially improve therapeutic management. Notably, macrophages have also been harnessed to deliver drugs. Noninvasive in vivo molecular imaging of macrophages is crucial for investigating intricate cellular processes, comprehending the underlying mechanisms of diseases, tracking cells and EVs' migration, and devising macrophage-dependent drug-delivery systems in living organisms. Thus, in vivo imaging of macrophages has become an indispensable tool in biomedical research. The integration of multimodal imaging approaches and the continued development of novel contrast agents hold promise for overcoming current limitations and expanding the applications of macrophage imaging. This study comprehensively reviews several methods for labeling macrophages and various imaging modalities, assessing the merits and drawbacks of each approach. The review concludes by offering insights into the applicability of molecular imaging techniques for real time monitoring of macrophages in preclinical and clinical scenarios.
Collapse
Affiliation(s)
- Prakash Gangadaran
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu, 41944, Korea
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, 41944, Korea
- Cardiovascular Research Institute, Kyungpook National University, Daegu, 41944, Republic of Korea
| | - Akanksha Onkar
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Ramya Lakshmi Rajendran
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu, 41944, Korea
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, 41944, Korea
- Cardiovascular Research Institute, Kyungpook National University, Daegu, 41944, Republic of Korea
| | - Anshika Goenka
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, 30322, USA
| | - Ji Min Oh
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, 41944, Korea
- Cardiovascular Research Institute, Kyungpook National University, Daegu, 41944, Republic of Korea
| | - Fatima Khan
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - ArulJothi Kandasamy Nagarajan
- Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu, 603203, Tamilnadu, India
| | - Sathish Muthu
- Department of Orthopaedics, Government Medical College, Tamil Nadu, 639004, Karur, India
- Department of Biotechnology, Faculty of Engineering, Karpagam Academy of Higher Education, Tamil Nadu, 641021, Coimbatore, India
| | - Anand Krishnan
- Precision Medicine and Integrated Nano-Diagnostics (P-MIND) Research Group, Office of the Dean, Faculty of Health Sciences, University of the Free State, Bloemfontein, 9300, South Africa
| | - Chae Moon Hong
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, 41944, Korea.
- Cardiovascular Research Institute, Kyungpook National University, Daegu, 41944, Republic of Korea.
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, 41944, Korea.
| | - Byeong-Cheol Ahn
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu, 41944, Korea.
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, 41944, Korea.
- Cardiovascular Research Institute, Kyungpook National University, Daegu, 41944, Republic of Korea.
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, 41944, Korea.
| |
Collapse
|
32
|
Yang Y, Wang J, Zhong Y, Tian M, Zhang H. Advances in Radionuclide-Labeled Biological Carriers for Tumor Imaging and Treatment. ACS APPLIED MATERIALS & INTERFACES 2025; 17:4316-4336. [PMID: 39792777 DOI: 10.1021/acsami.4c19059] [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: 01/12/2025]
Abstract
Biological carriers have emerged as significant tools to deliver radionuclides in nuclear medicine, providing a meaningful perspective for tumor imaging and treatment. Various radionuclide-labeled biological carriers have been developed to meet the needs of biomedical applications. This review introduces the principles of radionuclide-mediated imaging and therapy and the selected criteria of them, as well as a comprehensive description of the characteristics and functions of representative biological carriers including bacteria, cells, viruses, and their biological derivatives, emphasizing the labeled strategies of biological carriers combined with radionuclides. Subsequently, we in-depth introduce the application of radionuclide-labeled biological carriers in tumor imaging and treatment, including the imaging of the behaviors of biological carriers in vivo and tumor metastasis and the tumor treatment by radionuclide therapy, plus other strategies and radiation-induced photodynamic therapy. Finally, the challenges and prospects of radionuclide-labeled biological carriers are discussed to improve the shortcomings of this innovative platform and promote clinical transformation in the field of medical imaging.
Collapse
Affiliation(s)
- Yaozhang Yang
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
- Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, Zhejiang 310009, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, Zhejiang 310009, China
| | - Jing Wang
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
- Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, Zhejiang 310009, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, Zhejiang 310009, China
| | - Yan Zhong
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
- Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, Zhejiang 310009, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, Zhejiang 310009, China
| | - Mei Tian
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
- Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, Zhejiang 310009, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, Zhejiang 310009, China
- Human Phenome Institute, Fudan University, Shanghai 201203, China
| | - Hong Zhang
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
- Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, Zhejiang 310009, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, Zhejiang 310009, China
- College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou, Zhejiang 310014, China
- Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University, Hangzhou, Zhejiang 310014, China
| |
Collapse
|
33
|
An Y, Zhao F, Jia H, Meng S, Zhang Z, Li S, Zhao J. Inhibition of programmed cell death by melanoma cell subpopulations reveals mechanisms of melanoma metastasis and potential therapeutic targets. Discov Oncol 2025; 16:62. [PMID: 39832036 PMCID: PMC11747064 DOI: 10.1007/s12672-025-01789-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Accepted: 01/08/2025] [Indexed: 01/22/2025] Open
Abstract
Melanoma is an aggressive type of skin cancer that arises from melanocytes, the cells responsible for producing skin pigment. In contrast to non-melanoma skin cancers like basal cell carcinoma and squamous cell carcinoma, melanoma is more invasive. Melanoma was distinguished by its rapid progression, high metastatic potential, and significant resistance to conventional therapies. Although it accounted for a small proportion of skin cancer cases, melanoma accounts for the majority of deaths caused by skin cancer due to its ability to invade deep tissues, adapt to diverse microenvironments, and evade immune responses. These unique features highlighted the challenges of treating melanoma and underscored the importance of advanced tools, such as single-cell sequencing, to unravel its biology and develop personalized therapeutic strategies. Thus, we conducted a single-cell analysis of the cellular composition within melanoma tumor tissues and further subdivided melanoma cells into subpopulations. Through analyzing metabolic pathways, stemness genes, and transcription factors (TFs) among cells in different phases (G1, G2/M, and S) as well as between primary and metastatic foci cells, we investigated the specific mechanisms underlying melanoma metastasis. We also revisited the cellular stemness and temporal trajectories of melanoma cell subpopulations, identifying the core subpopulation as C0 SOD3 + Melanoma cells. Our findings revealed a close relationship between the pivotal C0 SOD3 + Melanoma cells subpopulation and oxidative pathways in metastatic tumor tissues. Additionally, we analyzed prognostically relevant differentially expressed genes (DEGs) within the C0 SOD3 + Melanoma cells subpopulation and built a predictive model associated with melanoma outcomes. We selected the gene IGF1 with the highest coefficient (coef) value for further analysis, and experimentally validated its essential function in the proliferation and invasive metastasis of melanoma. In immune infiltration analysis, we discovered the critical roles played by M1/M2 macrophages in melanoma progression and immune evasion. Furthermore, the development and progression of malignant melanoma were closely associated with various forms of programmed cell death (PCD), including apoptosis, autophagic cell death, ferroptosis, and pyroptosis. Melanoma cells often resisted cell death mechanisms, maintaining their growth by inhibiting apoptosis and evading autophagic cell death. Meanwhile, the induction of ferroptosis and pyroptosis was thought to trigger immune responses that helped suppress melanoma dissemination. A deeper understanding of the relationship between melanoma and PCD pathways provided a critical foundation for developing novel targeted therapies, with the potential to enhance melanoma treatment efficacy. These findings contributed to the development of novel prognostic models for melanoma and shed light on research directions concerning melanoma metastasis mechanisms and therapeutic targets.
Collapse
Affiliation(s)
- Yuepeng An
- The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Fu Zhao
- Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Hongling Jia
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shandong, China
| | - Siyu Meng
- Northeast International Hospital, Shenyang, 110180, China
| | - Ziwei Zhang
- Department of Plastic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| | - Shuxiao Li
- Department of Burns and Plastic Reconstructive Surgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, Guangxi Province, China.
- Life Science and Clinical Medicine Research Center, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, Guangxi Province, China.
| | - Jiusi Zhao
- The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, 150040, China.
| |
Collapse
|
34
|
Zhang Y, Wang W, Chen L, Wang H, Dong D, Zhu J, Guo Y, Zhou Y, Liu T, Fu W. Human adipose-derived multipotent stromal cells enriched with IL-10 modRNA improve diabetic wound healing: Trigger the macrophage phenotype shift. Bioeng Transl Med 2025; 10:e10711. [PMID: 39801749 PMCID: PMC11711206 DOI: 10.1002/btm2.10711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 07/15/2024] [Accepted: 07/23/2024] [Indexed: 01/16/2025] Open
Abstract
Diabetic wounds present a significant challenge in regenerative medicine due to impaired healing, characterized by prolonged inflammation and deficient tissue repair, primarily caused by a skewed pro-inflammatory macrophage phenotype. This study investigates the therapeutic potential of interleukin-10 (IL-10) chemically modified mRNA (modRNA)-enriched human adipose-derived multipotent stromal cells (hADSCs) in a well-established murine model of diabetic wounds. The modRNAs used in this study were chemically modified using N1-methylpseudouridine-5'-triphosphate (m1Ψ) by substituting uridine-5-triphosphate. In vitro experiments demonstrated that IL-10 modRNA-transfected hADSCs effectively modulated macrophage polarization towards an anti-inflammatory phenotype. In vivo experiments with a well-established murine model demonstrated that transplantation of hADSCsmodIL-10 on postoperative day 5 (POD5) significantly improved wound healing outcomes, including accelerated wound closure, enhanced re-epithelialization, promoted M2 polarization, improved collagen deposition, and increased neovascularization. This study concludes that IL-10 modRNA-enriched hADSCs offer a promising therapeutic approach for diabetic wound healing, with the timing of IL-10 administration playing a crucial role in its effectiveness. These cells modulate macrophage polarization and promote tissue repair, demonstrating their potential for improving the management of diabetic wounds.
Collapse
Affiliation(s)
- Yuxin Zhang
- Shanghai Key Laboratory of Clinical Geriatric MedicineHuadong HospitalShanghaiChina
- Department of Plastic Surgery, Huadong Hospital, School of MedicineFudan UniversityShanghaiChina
| | - Wei Wang
- Shanghai Key Laboratory of Clinical Geriatric MedicineHuadong HospitalShanghaiChina
- Department of Plastic Surgery, Huadong Hospital, School of MedicineFudan UniversityShanghaiChina
| | - Liang Chen
- Department of Plastic Surgery, Huadong Hospital, School of MedicineFudan UniversityShanghaiChina
| | - Heng Wang
- Department of Plastic Surgery, Huadong Hospital, School of MedicineFudan UniversityShanghaiChina
| | - Dong Dong
- Department of Plastic Surgery, Huadong Hospital, School of MedicineFudan UniversityShanghaiChina
| | - Jingjing Zhu
- Department of Plastic Surgery, Huadong Hospital, School of MedicineFudan UniversityShanghaiChina
| | - Yu Guo
- Department of Plastic Surgery, Huadong Hospital, School of MedicineFudan UniversityShanghaiChina
| | - Yiqun Zhou
- Department of Plastic Surgery, Huadong Hospital, School of MedicineFudan UniversityShanghaiChina
| | - Tianyi Liu
- Shanghai Key Laboratory of Clinical Geriatric MedicineHuadong HospitalShanghaiChina
- Department of Plastic Surgery, Huadong Hospital, School of MedicineFudan UniversityShanghaiChina
| | - Wei Fu
- Institute of Pediatric Translational Medicine, Shanghai Institute of Pediatric Congenital Heart Disease, Shanghai Children's Medical Center, School of MedicineShanghai Jiao Tong UniversityShanghaiChina
| |
Collapse
|
35
|
Chen C, Yang J, Shang R, Tang Y, Cai X, Chen Y, Liu Z, Hu W, Zhang W, Zhang X, Huang Y, Hu X, Yin W, Lu Q, Sheng H, Fan D, Ju Z, Luo G, He W. Orchestration of Macrophage Polarization Dynamics by Fibroblast-Secreted Exosomes during Skin Wound Healing. J Invest Dermatol 2025; 145:171-184.e6. [PMID: 38838771 DOI: 10.1016/j.jid.2024.05.007] [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: 02/12/2024] [Revised: 04/30/2024] [Accepted: 05/04/2024] [Indexed: 06/07/2024]
Abstract
Macrophages undertake pivotal yet dichotomous functions during skin wound healing, mediating both early proinflammatory immune activation and late anti-inflammatory tissue remodeling processes. The timely phenotypic transition of macrophages from inflammatory M1 to proresolving M2 activation states is essential for efficient healing. However, the endogenous mechanisms calibrating macrophage polarization in accordance with the evolving tissue milieu remain undefined. In this study, we reveal an indispensable immunomodulatory role for fibroblast-secreted exosomes in directing macrophage activation dynamics. Fibroblast-derived exosomes permitted spatiotemporal coordination of macrophage phenotypes independent of direct intercellular contact. Exosomes enhanced macrophage sensitivity to both M1 and M2 polarizing stimuli, yet they also accelerated timely switching from M1 to M2 phenotypes. Exosome inhibition dysregulated macrophage responses, resulting in aberrant inflammation and impaired healing, whereas provision of exogenous fibroblast-derived exosomes corrected defects. Topical application of fibroblast-derived exosomes onto chronic diabetic wounds normalized dysregulated macrophage activation to resolve inflammation and restore productive healing. Our findings elucidate fibroblast-secreted exosomes as remote programmers of macrophage polarization that calibrate immunological transitions essential for tissue repair. Harnessing exosomes represents a previously unreported approach to steer productive macrophage activation states with immense therapeutic potential for promoting healing in chronic inflammatory disorders.
Collapse
Affiliation(s)
- Cheng Chen
- Institute of Burn Research, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China; Chongqing Key Laboratory for Disease Proteomics, Chongqing, China
| | - Jiacai Yang
- Institute of Burn Research, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China; Chongqing Key Laboratory for Disease Proteomics, Chongqing, China
| | - Ruoyu Shang
- Institute of Burn Research, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China; Chongqing Key Laboratory for Disease Proteomics, Chongqing, China
| | - Yuanyang Tang
- Department of Infectious Diseases, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xin Cai
- Institute of Burn Research, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China; Chongqing Key Laboratory for Disease Proteomics, Chongqing, China
| | - Yunxia Chen
- Institute of Burn Research, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China; Chongqing Key Laboratory for Disease Proteomics, Chongqing, China
| | - Zhihui Liu
- Institute of Burn Research, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China; Chongqing Key Laboratory for Disease Proteomics, Chongqing, China
| | - Wengang Hu
- Institute of Burn Research, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China; Chongqing Key Laboratory for Disease Proteomics, Chongqing, China
| | - Weiguang Zhang
- Department of Intensive Care, Southwest Hospital, Army Medical University, Chongqing, China
| | - Xiaorong Zhang
- Institute of Burn Research, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China; Chongqing Key Laboratory for Disease Proteomics, Chongqing, China
| | - Yong Huang
- Institute of Burn Research, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China; Chongqing Key Laboratory for Disease Proteomics, Chongqing, China
| | - Xiaohong Hu
- Institute of Burn Research, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China; Chongqing Key Laboratory for Disease Proteomics, Chongqing, China
| | - Wenjing Yin
- Institute of Burn Research, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China; Chongqing Key Laboratory for Disease Proteomics, Chongqing, China; Academy of Biological Engineering, Chongqing University, Chongqing, China
| | - Qudong Lu
- Department of Urology, Army 73rd Group Military Hospital, Xiamen, China
| | - Hao Sheng
- Department of Urology, The Second Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Dejiang Fan
- Institute of Burn Research, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China; Chongqing Key Laboratory for Disease Proteomics, Chongqing, China
| | - Zhenyu Ju
- Key Laboratory of Regenerative Medicine of Ministry of Education, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Institute of Aging and Regenerative Medicine, Jinan University, Guangzhou, China
| | - Gaoxing Luo
- Institute of Burn Research, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China; Chongqing Key Laboratory for Disease Proteomics, Chongqing, China.
| | - Weifeng He
- Institute of Burn Research, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China; Chongqing Key Laboratory for Disease Proteomics, Chongqing, China.
| |
Collapse
|
36
|
Lin W, Shen C, Li M, Ma S, Liu C, Huang J, Ren Z, Yang Y, Zhao M, Xie Q, Guo S, Wang W, Wang K, Ma Q, Jiang Y, Zheng J, Liao Y. Programmable Macrophage Vesicle Based Bionic Self-Adjuvanting Vaccine for Immunization against Monkeypox Virus. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2408608. [PMID: 39513669 PMCID: PMC11714231 DOI: 10.1002/advs.202408608] [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: 07/25/2024] [Revised: 11/01/2024] [Indexed: 11/15/2024]
Abstract
The emergence of monkeypox has become a global health threat after the COVID-19 pandemic. Due to the lack of available specifically treatment against MPV, developing an available vaccine is thus the most prospective and urgent strategy. Herein, a programmable macrophage vesicle based bionic self-adjuvanting vaccine (AM@AEvs-PB) is first developed for defending against monkeypox virus (MPV). Based on MPV-related antigen-stimulated macrophage-derived vesicles, the nanovaccine is constructed by loading the mature virion (MV)-related intracellular protein (A29L/M1R) and simultaneously modifying with the enveloped virion (EV) antigen (B6R), enabling them to effectively promote antigen presentation and enhance adaptive immune through self-adjuvant strategy. Owing to the synergistic properties of bionic vaccine coloaded MV and EV protein in defensing MPV, the activation ratio of antigen-presenting cells is nearly four times than that of single antigen in the same dose, resulting in stronger immunity in host. Notably, intramuscular injection uptake of AM@AEvs-PB demonstrated vigorous immune-protective effects in the mouse challenge attempt, offering a promising strategy for pre-clinical monkeypox vaccine development.
Collapse
Affiliation(s)
- Weiqiang Lin
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Key Laboratory of Vascular Injury and Repair ResearchNingxia Medical UniversityYinchuan750004P. R. China
- Institute for Engineering MedicineKunming Medical UniversityKunming650500P. R. China
| | - Chenguang Shen
- BSL‐3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public HealthSouthern Medical UniversityGuangzhou510515P. R. China
| | - Mengjun Li
- BSL‐3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public HealthSouthern Medical UniversityGuangzhou510515P. R. China
| | - Shengchao Ma
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Key Laboratory of Vascular Injury and Repair ResearchNingxia Medical UniversityYinchuan750004P. R. China
| | - Chenxin Liu
- School of Laboratory Medicine and BiotechnologySouthern Medical UniversityGuangzhou510515P. R. China
| | - Jialin Huang
- Molecular Diagnosis and Treatment Center for Infectious DiseasesDermatology Hospital, Southern Medical UniversityGuangzhou510091P. R. China
| | - Zuning Ren
- BSL‐3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public HealthSouthern Medical UniversityGuangzhou510515P. R. China
| | - Yuechao Yang
- Molecular Diagnosis and Treatment Center for Infectious DiseasesDermatology Hospital, Southern Medical UniversityGuangzhou510091P. R. China
| | - Minghai Zhao
- Molecular Diagnosis and Treatment Center for Infectious DiseasesDermatology Hospital, Southern Medical UniversityGuangzhou510091P. R. China
| | - Qiulin Xie
- Molecular Diagnosis and Treatment Center for Infectious DiseasesDermatology Hospital, Southern Medical UniversityGuangzhou510091P. R. China
| | - Shuang Guo
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Key Laboratory of Vascular Injury and Repair ResearchNingxia Medical UniversityYinchuan750004P. R. China
| | - Wei Wang
- BSL‐3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public HealthSouthern Medical UniversityGuangzhou510515P. R. China
| | - Kaiyuan Wang
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and EngineeringNational University of SingaporeSingapore119074Singapore
| | - Qiang Ma
- School of Laboratory Medicine and BiotechnologySouthern Medical UniversityGuangzhou510515P. R. China
| | - Yideng Jiang
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Key Laboratory of Vascular Injury and Repair ResearchNingxia Medical UniversityYinchuan750004P. R. China
| | - Judun Zheng
- Molecular Diagnosis and Treatment Center for Infectious DiseasesDermatology Hospital, Southern Medical UniversityGuangzhou510091P. R. China
| | - Yuhui Liao
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Key Laboratory of Vascular Injury and Repair ResearchNingxia Medical UniversityYinchuan750004P. R. China
- Institute for Engineering MedicineKunming Medical UniversityKunming650500P. R. China
| |
Collapse
|
37
|
Liu Y, Qin J, Li X, Wu G. Oxysterols in tumor immune microenvironment (TIME). J Steroid Biochem Mol Biol 2025; 245:106634. [PMID: 39551164 DOI: 10.1016/j.jsbmb.2024.106634] [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: 07/29/2024] [Revised: 10/22/2024] [Accepted: 11/09/2024] [Indexed: 11/19/2024]
Abstract
Oxysterols are compounds generated through oxidative reactions involving cholesterol and other steroid molecules. They play a crucial role in the tumor immune microenvironment by interacting with molecules such as the cell membrane receptor EBI2 and nuclear receptors like LXR and PXR. This interaction regulates immune cell signaling pathways, affecting proliferation, apoptosis, migration, and invasion in tumor-related processes. Activating these receptors alters the function and behavior of immune cells-such as macrophages, T cells, and dendritic cells-within the tumor microenvironment, thus promoting or inhibiting tumor development. Certain oxidized steroids can increase both the number and activation of infiltrating T cells, synergizing with anti-PD-1 to enhance anti-tumor efficacy. An in-depth study of the biological mechanisms of oxidized sterols will not only enhance our understanding of the complexity of the tumor immune microenvironment but may also reveal new therapeutic targets, providing innovative strategies for tumor immunotherapy.
Collapse
Affiliation(s)
- Yuanxin Liu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| | - Jie Qin
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| | - Xiaorui Li
- Department of Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang 110042, China.
| | - Guangzhen Wu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| |
Collapse
|
38
|
Lu B, Chai L, Zhang Z, Zhao G, Shao Y, Zheng Y, Jin X, Zheng J, Chai D, Ding J. Co-immunization with IFI35 enhances the therapeutic effect of an adenovirus vaccine against renal carcinoma. Int J Biol Macromol 2025; 286:138515. [PMID: 39647736 DOI: 10.1016/j.ijbiomac.2024.138515] [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: 09/22/2024] [Revised: 12/04/2024] [Accepted: 12/05/2024] [Indexed: 12/10/2024]
Abstract
Interferon-induced protein 35 (IFI35), an immunomodulator, is highly expressed in tumor cells, yet its role in enhancing tumor vaccine efficacy remains unclear. In this study, an adenovirus (Ad) vaccine encoding dual targets, IFI35 and carbonic anhydrase IX (CAIX), was developed for renal carcinoma treatment. Co-immunization with Ad-IFI35/CAIX effectively inhibited tumor growth in a subcutaneous model and significantly increased the infiltration of CD8+ T cells and dendritic cells (DCs). Furthermore, Ad-IFI35/CAIX administration induced strong cytotoxic T lymphocyte (CTL) responses and expanded multifunctional CD8+ T cell populations. Depletion of CD8+ T cells abolished the vaccine's tumor regression effects, confirming that its therapeutic effect relies on CD8+ T cell-mediated immunity. In addition, Ad-IFI35/CAIX treatment enhanced the induction of memory CTL responses, effectively suppressing the growth of tumors implanted contralaterally. The Ad-IFI35/CAIX vaccine also elicited a strong CD8+ T cell-mediated immunity against tumor metastasis and growth in lung metastasis and orthotopic renal carcinoma models. These results indicate that the Ad vaccine dual targeting IFI35 and CAIX is a potential strategy for renal carcinoma treatment.
Collapse
Affiliation(s)
- Bowen Lu
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Leizi Chai
- Department of Traumatic Orthopaedics, People's Hospital of Bozhou City, Bozhou Hospital of Anhui Medical University, Bozhou, Anhui 236000, China
| | - Zichun Zhang
- Department of Urology, The Yancheng Clinical College of Xuzhou Medical University, Yancheng City No. 1 People's Hospital, Yancheng, Jiangsu 224001, China
| | - Guangya Zhao
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Yingxiang Shao
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Yanyan Zheng
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Xin Jin
- Department of Oncology, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou, Jiangsu 221009, China
| | - Junnian Zheng
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China.
| | - Dafei Chai
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China.
| | - Jiage Ding
- Department of Oncology, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou, Jiangsu 221009, China.
| |
Collapse
|
39
|
Jiang W, Xu S, Li P. SLC2A3 is a Potential Factor for Head and Neck Squamous Cancer Development through Tumor Microenvironment Alteration. Curr Gene Ther 2025; 25:157-177. [PMID: 38778609 PMCID: PMC11774314 DOI: 10.2174/0115665232291300240509104344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 03/13/2024] [Accepted: 03/27/2024] [Indexed: 05/25/2024]
Abstract
INTRODUCTION Tumor immunity has garnered increasing attention in cancer treatment and progression. However, there is still a challenge in understanding the mechanisms of specific molecules affecting the clinical prognosis and tumor microenvironment (TME). METHODS Here, we applied the ESTIMATE algorithm to calculate the immune and stromal scores in 504 HNSC cases from TCGA. Patients were grouped according to the median value of the immune and stromal. Clinicopathological characteristics and differentially expressed genes (DEG) were analyzed. Subsequently, LASSO, COX regression, survival analysis, and clinicopathological characteristics were conducted. Subsequently, SLC2A3 was determined as a predictive factor that high expression of SLC2A3 at the mRNA and protein levels predicted a worse clinical prognosis. GSEA25099 was utilized for external validation of immune infiltration, while tissue PCR, IHC, and Western Blot were used to confirm the expression levels of SLC2A3. RESULTS A series of immune-infiltration analyses showed that SLC2A3 expression was negatively correlated with CD8+ T cells, significantly affecting the survival prognosis of HNSC. In the GSEA analysis, the high expression of SLC2A3 was mainly enriched for immune-related biological processes. Meanwhile, high expression of SLC2A3 possessed higher TIDE scores and was also strongly positively correlated with a series of immune checkpoints affecting survival prognosis, thus causing greater susceptibility to immune escape. CONCLUSION Conclusively, SLC2A3 is a potential oncogene and factor of HNSC development, notably by an altered state of the immune microenvironment, immune-suppressive regulation, and immune escape.
Collapse
Affiliation(s)
- Wei Jiang
- Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
- College of Stomatology, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Sheng Xu
- Department of Dental Laboratory, Guangxi Medical University College of Stomatology, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Ping Li
- Department of Pathology, Guangxi Medical University College of Stomatology, Nanning, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
40
|
Cao Y, Yan W, Yi W, Yin Q, Li Y. Bioengineered therapeutic systems for improving antitumor immunity. Natl Sci Rev 2025; 12:nwae404. [PMID: 40114728 PMCID: PMC11925021 DOI: 10.1093/nsr/nwae404] [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: 08/22/2024] [Revised: 09/29/2024] [Accepted: 11/08/2024] [Indexed: 03/22/2025] Open
Abstract
Immunotherapy, a monumental advancement in antitumor therapy, still yields limited clinical benefits owing to its unguaranteed efficacy and safety. Therapeutic systems derived from cellular, bacterial and viral sources possess inherent properties that are conducive to antitumor immunotherapy. However, crude biomimetic systems have restricted functionality and may produce undesired toxicity. With advances in biotechnology, various toolkits are available to add or subtract certain properties of living organisms to create flexible therapeutic platforms. This review elaborates on the creation of bioengineered systems, via gene editing, synthetic biology and surface engineering, to enhance immunotherapy. The modifying strategies of the systems are discussed, including equipment for navigation and recognition systems to improve therapeutic precision, the introduction of controllable components to control the duration and intensity of treatment, the addition of immunomodulatory components to amplify immune activation, and the removal of toxicity factors to ensure biosafety. Finally, we summarize the advantages of bioengineered immunotherapeutic systems and possible directions for their clinical translation.
Collapse
Affiliation(s)
- Ying Cao
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Life Sciences, Jilin University, Changchun 130012, China
| | - Wenlu Yan
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wenzhe Yi
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qi Yin
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai 264000, China
| | - Yaping Li
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai 264000, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264000, China
| |
Collapse
|
41
|
Chen S, Liu J, Zhu L. M2-like macrophage-derived exosomes inhibit osteoclastogenesis via releasing miR-1227-5p. Immunobiology 2025; 230:152861. [PMID: 39700638 DOI: 10.1016/j.imbio.2024.152861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 11/27/2024] [Accepted: 12/05/2024] [Indexed: 12/21/2024]
Abstract
Macrophages play a pivotal role in regulating inflammatory response in periodontitis, a condition characterized by excessive osteoclast differentiation. This study aimed to investigate whether exosomes derived from M2 macrophages regulate osteoclast differentiation and to identify the underlying molecular mechanisms. Exosomes were isolated from M2 macrophages and used to treat osteoclasts. Osteoclastogenesis was assessed using tartrate-resistant acid phosphatase staining and reverse transcription-quantitative polymerase chain reaction (RT-qPCR). The molecular mechanism was evaluated using microarray analysis, RT-qPCR, dual-luciferase reporter analysis, and RNA pull-down assay. The results showed that exosomes from M2 macrophages inhibited receptor activator of nuclear factor κ-B ligand (RANKL)-induced osteoclast differentiation. Additionally, miR-1227-5p expression in osteoclasts was increased after treatment with exosomes, and inhibition of miR-1227-5p counteracted the suppressive effects of exosomes on osteoclastogenesis. Moreover, OSCAR is a target of miR-1227-5p. In conclusion, exosomal miR-1227-5p suppresses osteoclast differentiation, potentially via targeting OSCAR. These findings provide new insights into the pathogenesis of periodontitis.
Collapse
Affiliation(s)
- Shan Chen
- Department of Periodontology, Changsha Stomatological Hospital, No. 389, Youyi Road, Tianxin District, Changsha 410004, China
| | - Jian Liu
- Department of Periodontology, Changsha Stomatological Hospital, No. 389, Youyi Road, Tianxin District, Changsha 410004, China
| | - Lilei Zhu
- Department of Periodontology, Changsha Stomatological Hospital, No. 389, Youyi Road, Tianxin District, Changsha 410004, China.
| |
Collapse
|
42
|
Lehrich BM, Delgado ER. Lipid Nanovesicle Platforms for Hepatocellular Carcinoma Precision Medicine Therapeutics: Progress and Perspectives. Organogenesis 2024; 20:2313696. [PMID: 38357804 PMCID: PMC10878025 DOI: 10.1080/15476278.2024.2313696] [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: 07/06/2023] [Revised: 01/04/2024] [Accepted: 01/30/2024] [Indexed: 02/16/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-related mortality globally. HCC is highly heterogenous with diverse etiologies leading to different driver mutations potentiating unique tumor immune microenvironments. Current therapeutic options, including immune checkpoint inhibitors and combinations, have achieved limited objective response rates for the majority of patients. Thus, a precision medicine approach is needed to tailor specific treatment options for molecular subsets of HCC patients. Lipid nanovesicle platforms, either liposome- (synthetic) or extracellular vesicle (natural)-derived present are improved drug delivery vehicles which may be modified to contain specific cargos for targeting specific tumor sites, with a natural affinity for liver with limited toxicity. This mini-review provides updates on the applications of novel lipid nanovesicle-based therapeutics for HCC precision medicine and the challenges associated with translating this therapeutic subclass from preclinical models to the clinic.
Collapse
Affiliation(s)
- Brandon M. Lehrich
- Division of Experimental Pathology, Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Medical Scientist Training Program, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Evan R. Delgado
- Division of Experimental Pathology, Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
43
|
Tan J, Feng L, Ragavan ND, Chai Theam O, Li X. The promotive effect of Caspase-11 overexpression in a rat model of chronic kidney disease and the therapeutic efficacy of exosome-delivered siRNA in inhibiting Caspase-11. Biochem Biophys Res Commun 2024; 741:151013. [PMID: 39591906 DOI: 10.1016/j.bbrc.2024.151013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 11/06/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024]
Abstract
This study investigates the role of Caspase-11 in Chronic Kidney Disease (CKD) and examines the therapeutic potential of inhibiting Caspase-11 using exosome-mediated siRNA. We established a CKD rat model and analyzed the expression of Caspase-11 through immunohistochemistry. The study involved overexpressing Caspase-11 using an adeno-associated virus (AAV) and constructing exosomes loaded with siRNA targeting Caspase-11 (exo-si-Caspase-11). Renal tissue damage and fibrosis were assessed using H&E staining, Masson's trichrome, TUNEL assay, and Sirius Red staining. Additionally, urinary protein and blood urea nitrogen (BUN) levels were measured, alongside analyses of serum calcium and phosphorus levels. H&E staining was performed to evaluate the effects of exo-si-Caspase-11 on damage to the heart, liver, spleen, and lungs. The results showed that the CKD model group experienced significant weight loss, increased blood pressure, and elevated Caspase-11 expression. AAV-mediated Caspase-11 overexpression led to substantial renal fibrosis, increased apoptosis, and elevated urinary protein and BUN levels. Additionally, the group with Caspase-11 overexpression exhibited elevated serum calcium and phosphorus levels. Conversely, treatment with exo-si-Caspase-11 reduced these pathological changes in renal tissue without causing damage to other major organs. These findings suggest that exosome-mediated siRNA delivery targeting Caspase-11 is an effective therapeutic strategy for CKD.
Collapse
MESH Headings
- Animals
- Renal Insufficiency, Chronic/therapy
- Renal Insufficiency, Chronic/genetics
- Renal Insufficiency, Chronic/pathology
- Renal Insufficiency, Chronic/metabolism
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Exosomes/metabolism
- Exosomes/genetics
- Disease Models, Animal
- Male
- Rats
- Rats, Sprague-Dawley
- Caspases, Initiator/genetics
- Caspases, Initiator/metabolism
- Kidney/pathology
- Kidney/metabolism
- Apoptosis/genetics
- Fibrosis
- Dependovirus/genetics
- Caspase 12
Collapse
Affiliation(s)
- Junhua Tan
- Department of Nephrology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, 533000, China; Faculty of Medicine, MAHSA University, Jalan SP 2, Bandar Saujana Putra, 42610 Jenjarom, Selangor, Malaysia; Key Laboratory of Medical Research Basic Guaranteefor Immune-Related Diseases Research of Guangxi (Cultivation), Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, 533000, China.
| | - Liyin Feng
- Graduate School of Youjiang Medical College for Nationalities, Baise, Guangxi, 533000, China
| | - Nanthiney Devi Ragavan
- School of Bioscience, Faculty of Pharmacy and Biomedical Sciences, MAHSA University, Jalan SP2, Bandar Saujana Putra, 42610, Jenjarom, Selangor, Malaysia
| | - Ooi Chai Theam
- Departmental of Preclinical Science,Faculty of Dentistry, MAHSA University Jalan SP 2, Bandar Saujana Putra, 42610, Jenjarom, Selangor, Malaysia.
| | - Xuebin Li
- Department of Neurology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, 533000, China; Department of Neurology, Youjiang Medical University for Nationalities, Baise, Guangxi, 533000, China.
| |
Collapse
|
44
|
Wang D, Han X, Liu HL. The role and research progress of tumor-associated macrophages in cervical cancer. Am J Cancer Res 2024; 14:5999-6011. [PMID: 39803646 PMCID: PMC11711540 DOI: 10.62347/ffxl7288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 12/17/2024] [Indexed: 01/16/2025] Open
Abstract
Tumor-associated macrophages (TAMs) are important immune cells in the tumor micro-environment (TME) and play a key role in the occurrence and development of cervical cancer. Besides, targeting TAMs can significantly inhibit cervical cancer tumor growth, invasion, metastasis, and angiogenesis as well as affect immune regulation. This review summarizes the correlation between TAM and tumors, the mechanism of action of TAM in cervical cancer, and the potential application of TAM in the treatment of cervical cancer. Therefore, this study may provide new ideas and targets for the development of further treatment strategies for cervical cancer patients.
Collapse
Affiliation(s)
- Dan Wang
- Department of First Clinical Medical College, Gansu University of Chinese MedicineLanzhou, Gansu, China
| | - Xue Han
- Department of Gynecology, Gansu Provincial People’s HospitalLanzhou, Gansu, China
| | - Hui-Ling Liu
- Department of Gynecology, Gansu Provincial People’s HospitalLanzhou, Gansu, China
| |
Collapse
|
45
|
Zhou F, Tao J, Gou H, Liu S, Yu D, Zhang J, Ji J, Lin N, Wang Y. FSTL1 sustains glioma stem cell stemness and promotes immunosuppressive macrophage polarization in glioblastoma. Cancer Lett 2024; 611:217400. [PMID: 39722404 DOI: 10.1016/j.canlet.2024.217400] [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: 10/01/2024] [Revised: 10/27/2024] [Accepted: 12/14/2024] [Indexed: 12/28/2024]
Abstract
Tumor-associated macrophages (TAMs) within the tumor microenvironment (TME) play a crucial role in glioblastoma (GBM) progression by interacting with glioma stem cells (GSCs). These interactions lead to the polarization of TAMs toward an M2 phenotype, which, in turn, enhances the stem-like traits and malignant progression of GSCs. Our study shows that FSTL1, a protein released by GSCs, is significantly elevated in gliomas and linked to the progression of the disease. By suppressing FSTL1 in a mouse model, we observed reduced tumor growth and a decrease in M2 macrophages. In vitro studies show that FSTL1 from GSCs promotes M2 polarization and infiltration. Importantly, GSCs utilize autocrine FSTL1 to interact with TLR2, which inhibits the endocytosis-lysosomal degradation pathway mediated by EGFR, resulting in the activation of the PI3K-AKT signaling pathway that is critical for maintaining their self-renewal. These findings underscore the importance of FSTL1 in GSC maintenance and M2 macrophage polarization, suggesting that interventions targeting the FSTL1/TLR2 pathway could provide a novel therapeutic approach for GBM patients.
Collapse
Affiliation(s)
- Fengqi Zhou
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China; Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jincheng Tao
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Huiqing Gou
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
| | - Shuheng Liu
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Dong Yu
- Department of Neurosurgery, The Affiliated Chuzhou Hospital of Anhui Medical University, The First People's Hospital of Chuzhou, Chuzhou, Anhui, China
| | - Junxia Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jianxiong Ji
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| | - Ning Lin
- Department of Neurosurgery, The Affiliated Chuzhou Hospital of Anhui Medical University, The First People's Hospital of Chuzhou, Chuzhou, Anhui, China.
| | - Yingyi Wang
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
46
|
Wang L, Liu H, Chen G, Wu Q, Xu S, Zhou Q, Zhao Y, Wang Q, Yan T, Cheng X. Bubble Ticket Trip: Exploring the Mechanism of miRNA Sorting into Exosomes and Maintaining the Stability of Tumor Microenvironment. Int J Nanomedicine 2024; 19:13671-13685. [PMID: 39723172 PMCID: PMC11669276 DOI: 10.2147/ijn.s498599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 12/14/2024] [Indexed: 12/28/2024] Open
Abstract
Exosomes are vesicles ranging from 30 to 100 nanometers in size that show great potential as carriers for therapeutic uses and drug delivery. Enriching a specific set of miRNAs in exosomes emphasizes the existence of particular sorting mechanisms that manage the targeted cargo packaging. The molecular mechanism for miRNA sorting has not been understood. It is crucial to understand the mechanism of exosome encapsulation to develop its therapeutic potential. In this review, we will explore the particular processes through which exosomes naturally encapsulate miRNA, as well as discuss the effect on tumors after encapsulation of miRNAs. We also summarize the effects of targeted drug delivery using genetic engineering and chemical methods to modify exosome-encapsulated miRNA. Finally, gaining insight into how exosome cargo is sorted could be applied in clinical settings for precise drug delivery and to hinder the progression of diseases.
Collapse
Affiliation(s)
- Lu Wang
- Key Laboratory of Cellular Physiology of the Ministry of Education, & Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi Province, 030001, People's Republic of China
| | - Huijuan Liu
- Key Laboratory of Cellular Physiology of the Ministry of Education, & Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi Province, 030001, People's Republic of China
| | - Guohui Chen
- Key Laboratory of Cellular Physiology of the Ministry of Education, & Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi Province, 030001, People's Republic of China
| | - Qinglu Wu
- Key Laboratory of Cellular Physiology of the Ministry of Education, & Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi Province, 030001, People's Republic of China
| | - Songrui Xu
- Key Laboratory of Cellular Physiology of the Ministry of Education, & Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi Province, 030001, People's Republic of China
| | - Qichao Zhou
- Key Laboratory of Cellular Physiology of the Ministry of Education, & Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi Province, 030001, People's Republic of China
| | - Yadong Zhao
- Key Laboratory of Cellular Physiology of the Ministry of Education, & Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi Province, 030001, People's Republic of China
| | - Qiaorong Wang
- Key Laboratory of Cellular Physiology of the Ministry of Education, & Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi Province, 030001, People's Republic of China
| | - Ting Yan
- Key Laboratory of Cellular Physiology of the Ministry of Education, & Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi Province, 030001, People's Republic of China
| | - Xiaolong Cheng
- Key Laboratory of Cellular Physiology of the Ministry of Education, & Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi Province, 030001, People's Republic of China
| |
Collapse
|
47
|
Xu M, Hu Y, Wu J, Liu J, Pu K. Sonodynamic Nano-LYTACs Reverse Tumor Immunosuppressive Microenvironment for Cancer Immunotherapy. J Am Chem Soc 2024; 146:34669-34680. [PMID: 39644208 DOI: 10.1021/jacs.4c13022] [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: 12/09/2024]
Abstract
Extracellular and transmembrane proteins, which account for the products of approximately 40% of all protein-encoding genes in tumors, play a crucial role in shaping the tumor immunosuppressive microenvironment (TIME). While protein degradation therapy has been applied to membrane proteins of cancer cells, it has rarely been extended to immune cells. We herein report a polymeric nanolysosome targeting chimera (nano-LYTAC) that undergoes membrane protein degradation on M2 macrophages and generates a sonodynamic effect for combinational cancer immunotherapy. Nano-LYTAC is found to have higher degradation efficacy to the interleukin 4 receptor (IL-4R) compared to traditional inhibitors. More importantly, it is revealed that the effect of nano-LYTAC on the function of the M2 macrophage is concentration-dependent: downregulating CD206 expression and interleukin 10 (IL-10) secretion from M2 macrophages at low concentration, while triggering their apoptosis at high concentration. Moreover, nano-LYTAC is found to possess long tumor retention (>48 h), allowing for multiple sonodynamic treatments with a single dose. Such a synergistic sonodynamic immunotherapy mediated by nano-LYTAC effectively reprograms the TIME via inhibiting the functions of M2 macrophages and regulatory T cells (Tregs), as well as promoting the maturation of dendritic cells (DCs) and tumor infiltration of T effector cells (Teffs), completely suppressing tumor growth, inhibiting pulmonary metastasis, and preventing recurrence under preclinical animal models.
Collapse
Affiliation(s)
- Mengke Xu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore 637457
| | - Yuxuan Hu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore 637457
| | - Jiayan Wu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore 637457
| | - Jing Liu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore 637457
| | - Kanyi Pu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore 637457
- Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive, Singapore 636921
| |
Collapse
|
48
|
Chen H, Lin Y, Chen J, Luo X, Kan Y, He Y, Zhu R, Jin J, Li D, Wang Y, Han Z. Targeting caspase-8: a new strategy for combating hepatocellular carcinoma. Front Immunol 2024; 15:1501659. [PMID: 39726605 PMCID: PMC11669555 DOI: 10.3389/fimmu.2024.1501659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 11/29/2024] [Indexed: 12/28/2024] Open
Abstract
Hepatocellular carcinoma (HCC) represents the most prevalent form of primary liver cancer and has a high mortality rate. Caspase-8 plays a pivotal role in an array of cellular signaling pathways and is essential for the governance of programmed cell death mechanisms, inflammatory responses, and the dynamics of the tumor microenvironment. Dysregulation of caspase-8 is intricately linked to the complex biological underpinnings of HCC. In this manuscript, we provide a comprehensive review of the regulatory roles of caspase-8 in apoptosis, necroptosis, pyroptosis, and PANoptosis, as well as its impact on inflammatory reactions and the intricate interplay with critical immune cells within the tumor microenvironment, such as tumor-associated macrophages, T cells, natural killer cells, and dendritic cells. Furthermore, we emphasize how caspase-8 plays pivotal roles in the development, progression, and drug resistance observed in HCC, and explore the potential of targeting caspase-8 as a promising strategy for HCC treatment.
Collapse
Affiliation(s)
- Haoran Chen
- Department of General Surgery, Chengdu Xinhua Hospital Affiliated to North Sichuan Medical College, Chengdu, China
| | - Yumeng Lin
- Health Management Center, Nanjing Tongren Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Jie Chen
- Department of General Surgery, Chengdu Xinhua Hospital Affiliated to North Sichuan Medical College, Chengdu, China
| | - Xuemei Luo
- Department of General Surgery, Chengdu Xinhua Hospital Affiliated to North Sichuan Medical College, Chengdu, China
| | - Yubo Kan
- Sichuan Provincial Woman’s and Children’s Hospital/The Affiliated Women’s and Children’s Hospital of Chengdu Medical College, Chengdu, China
| | - Yuqi He
- Department of Blood Transfusion, Lu’an People’s Hospital, the Affiliated Hospital of Anhui Medical University, Lu’an, China
| | - Renhe Zhu
- Department of Blood Transfusion, Lu’an People’s Hospital, the Affiliated Hospital of Anhui Medical University, Lu’an, China
| | - Jiahui Jin
- Department of gastroenterology, Baoji Central Hospital, Baoji, China
| | - Dongxuan Li
- Department of General Surgery, Chengdu Xinhua Hospital Affiliated to North Sichuan Medical College, Chengdu, China
| | - Yi Wang
- Department of General Surgery, Chengdu Xinhua Hospital Affiliated to North Sichuan Medical College, Chengdu, China
| | - Zhongyu Han
- Department of General Surgery, Chengdu Xinhua Hospital Affiliated to North Sichuan Medical College, Chengdu, China
| |
Collapse
|
49
|
Chen H, Han Z, Ma Y, Meng Q. Advances in macrophage-derived exosomes as immunomodulators in disease progression and therapy. Int Immunopharmacol 2024; 142:113248. [PMID: 39321698 DOI: 10.1016/j.intimp.2024.113248] [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: 06/18/2024] [Revised: 09/20/2024] [Accepted: 09/20/2024] [Indexed: 09/27/2024]
Abstract
Most somatic cells secrete vesicles called exosomes, which contain a variety of biomolecules. Recent research indicates that macrophage-derived exosomes are strongly correlated with tumors, infectious diseases, chronic inflammation, and tissue fibrosis. Therefore, the purpose of this review is to delve into the mechanisms of pathological states and how macrophage-derived exosomes react to them. We also discuss the biological effects of exosomes and how they affect disease. In addition, we have examined the possible uses of exosomes in illness treatment, highlighting both the benefits and drawbacks of these applications.
Collapse
Affiliation(s)
- Huizhu Chen
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing Key Laboratory of Sports Injuries, Beijing 100191, China; Peking University First Hospital, Peking University Health Science Center, Beijing 100034, China; Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing 100191, China.
| | - Ziping Han
- Institute of Cerebrovascular Diseases Research and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China.
| | - Yong Ma
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing Key Laboratory of Sports Injuries, Beijing 100191, China; Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing 100191, China.
| | - Qingyang Meng
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing Key Laboratory of Sports Injuries, Beijing 100191, China; Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing 100191, China.
| |
Collapse
|
50
|
Feng X, Lai X, Zhou M, Bie J, Li T, Wang D, Chen S, Hu X, Wang C, Xu P. Targeting HLA-E in Lung Cancer: The Therapeutic Potential of IRF5-Engineered M1-Macrophage-Derived Exosomes. THE CLINICAL RESPIRATORY JOURNAL 2024; 18:e70035. [PMID: 39623605 PMCID: PMC11611755 DOI: 10.1111/crj.70035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 10/23/2024] [Accepted: 11/04/2024] [Indexed: 12/06/2024]
Abstract
Immunotherapy is a pivotal approach in the treatment of lung cancer. Although HLA-E is a potential target for tumor immunotherapy, its role in lung cancer remains unclear. Previous studies have identified the transcription factor IRF5 as a characteristic gene of M1-like macrophages, highlighting its crucial role in promoting antitumor immune responses. In this study, we developed an engineered M1-like macrophage exosomes expressing IRF5 (IRF5 M1-exos) and demonstrated their ability to inhibit proliferation, migration, and invasion of lung cancer cells. Moreover, our experiments using a nude mouse model revealed that IRF5 M1-exos exerted potent therapeutic effects by effectively suppressing tumor growth. Notably, the mechanism by which IRF5 exerts its antitumor function through HLA-E regulation in lung cancer has not been fully elucidated. Here, we identified HLA-E as a downstream target gene of IRF5 and demonstrated that the overexpression of HLA-E can counteract the tumor-promoting effects induced by si-IRF5 M1-exos. These results suggest that M1 macrophage-derived exosomes, enriched with the transcription factor IRF5, exhibit potent antitumor activity by up-regulating HLA-E in lung cancer cells. Therefore, IRF5 M1-exos represent an attractive therapeutic strategy for lung cancer.
Collapse
Affiliation(s)
- Xuqin Feng
- Department of OncologyBeijing Anzhen Nanchong Hospital, Capital Medical University (Nanchong Central Hospital), The Second Clinical Medical College of North Sichuan Medical CollegeNanchongSichuanChina
| | - Xiangyu Lai
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing Cancer HospitalChongqing Cancer InstituteChongqingChina
| | - Mingming Zhou
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing Cancer HospitalChongqing Cancer InstituteChongqingChina
| | - Jun Bie
- Department of OncologyBeijing Anzhen Nanchong Hospital, Capital Medical University (Nanchong Central Hospital), The Second Clinical Medical College of North Sichuan Medical CollegeNanchongSichuanChina
| | - Tingting Li
- Department of OncologyBeijing Anzhen Nanchong Hospital, Capital Medical University (Nanchong Central Hospital), The Second Clinical Medical College of North Sichuan Medical CollegeNanchongSichuanChina
| | - Dan Wang
- Department of OncologyBeijing Anzhen Nanchong Hospital, Capital Medical University (Nanchong Central Hospital), The Second Clinical Medical College of North Sichuan Medical CollegeNanchongSichuanChina
| | - Silin Chen
- Department of OncologyBeijing Anzhen Nanchong Hospital, Capital Medical University (Nanchong Central Hospital), The Second Clinical Medical College of North Sichuan Medical CollegeNanchongSichuanChina
| | - Xin Hu
- Department of OncologyBeijing Anzhen Nanchong Hospital, Capital Medical University (Nanchong Central Hospital), The Second Clinical Medical College of North Sichuan Medical CollegeNanchongSichuanChina
| | - Chunyu Wang
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing Cancer HospitalChongqing Cancer InstituteChongqingChina
| | - Peng Xu
- Department of Clinical LaboratoryBeibei Traditional Chinese Medical HospitalChongqingChina
| |
Collapse
|