1
|
Kong M, Li J, Tong N. The role of peripheral blood microRNAs in the pathogenesis and treatment response of age-related macular degeneration. Future Sci OA 2025; 11:2482499. [PMID: 40183456 PMCID: PMC11980467 DOI: 10.1080/20565623.2025.2482499] [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: 11/02/2024] [Accepted: 02/12/2025] [Indexed: 04/05/2025] Open
Abstract
Age-related macular degeneration is a leading cause of vision loss in aging populations, driven by complex interactions between genetic, environmental, and molecular factors. MicroRNAs have emerged as crucial regulators of cellular processes such as oxidative stress, inflammation, and angiogenesis, all of which contribute to AMD pathogenesis. This narrative review aims to summarize the involvement of peripheral blood microRNAs in the pathogenesis of AMD, focusing on key pathways such as oxidative stress, inflammation, and angiogenesis. Additionally, it explores their potential as biomarkers for predicting treatment response, particularly to anti-VEGF therapies. The potential of miRNAs as noninvasive biomarkers for early diagnosis and personalized treatment strategies is also explored, highlighting future directions for research.
Collapse
Affiliation(s)
- Meng Kong
- Department of Ophthalmology, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, Shandong, China
- School of Medicine, Qingdao University, Qingdao, Shandong, China
| | - Jingwen Li
- Department of Ophthalmology, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, Shandong, China
| | - Nianting Tong
- Department of Ophthalmology, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, Shandong, China
- School of Medicine, Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
2
|
Liu L, Li Y, Li B. Interactions between cancer cells and tumor-associated macrophages in tumor microenvironment. Biochim Biophys Acta Rev Cancer 2025; 1880:189344. [PMID: 40345263 DOI: 10.1016/j.bbcan.2025.189344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 05/02/2025] [Accepted: 05/05/2025] [Indexed: 05/11/2025]
Abstract
Tumor microenvironment (TME) refers to the local environment in which various cancer cells grow, encompassing tumor cells, adjacent non-tumor cells, and associated non-cellular elements, all of which collectively promote cancer occurrence and progression. As a principal immune component in the TME, tumor-associated macrophages (TAMs) exert a considerable influence on cancer behaviors via their interactions with cancer cells. The interactive loops between cancer cells and TAMs, including secretory factors derived from both cancer cells and TAMs, are crucial for the proliferation, stemness, drug resistance, invasion, migration, metastasis, and immune escape of various cancers. Cancer cells release paracrine proteins (HMGB1, AREG etc.), cytokines (IL-6, CCL2 etc.), RNAs (miR-21-5p, circPLEKHM1, LINC01812 etc.), and metabolites (lactic acid, succinate etc.) to regulate the polarization phenotype, mediator secretion and function of TAMs. In turn, mediators (TGF-β, IL-10, IL-6 etc.) from TAMs promote cancer progression. This review summarizes recent advancements in the interactive loops between cancer cells and TAMs in TME. Inhibiting the recruitment and M2 polarization of TAMs, reprogramming TAMs from M2 to M1 phenotype, blocking TAMs-mediated immunosuppression and immune escape, and combining with existing immunotherapy can target TAMs to overcome immunotherapy resistance in various cancers. The new breakthroughs lie in identifying effective targets for drug development, improving the drug delivery system to enhance the drug delivery efficiency, and adopting combined therapy. Interventions targeting secretory factors, cell surface receptors, intracellular signaling pathways, and metabolic modulation in the interactive loops between cancer cells and TAMs are expected to suppress cancer progression and improve therapeutic effects.
Collapse
Affiliation(s)
- Lu Liu
- Department of Oral Anatomy and Physiology, Jilin Provincial Key Laboratory of Oral Biomedical Engineering, Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Yafei Li
- Department of Oral Anatomy and Physiology, Jilin Provincial Key Laboratory of Oral Biomedical Engineering, Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Bo Li
- Department of Oral Anatomy and Physiology, Jilin Provincial Key Laboratory of Oral Biomedical Engineering, Hospital of Stomatology, Jilin University, Changchun 130021, China.
| |
Collapse
|
3
|
Safaei M, Rajabi SS, Tirgar M, Namdar N, Dalfardi M, Mohammadifar F, Goodarzi A, Farmani AR, Ramezani V, Abpeikar Z. Exosome-based approaches in cancer along with unlocking new insights into regeneration of cancer-prone tissues. Regen Ther 2025; 29:202-216. [DOI: https:/doi.org/10.1016/j.reth.2025.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2025] Open
|
4
|
Safaei M, Rajabi SS, Tirgar M, Namdar N, Dalfardi M, Mohammadifar F, Goodarzi A, Farmani AR, Ramezani V, Abpeikar Z. Exosome-based approaches in cancer along with unlocking new insights into regeneration of cancer-prone tissues. Regen Ther 2025; 29:202-216. [PMID: 40225049 PMCID: PMC11992408 DOI: 10.1016/j.reth.2025.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Revised: 03/01/2025] [Accepted: 03/18/2025] [Indexed: 04/15/2025] Open
Abstract
Most eukaryotic cells secrete extracellular vesicles called exosomes, which are involved in intercellular communication. Exosomes play a role in tumor development and metastasis by transporting bioactive chemicals from cancerous cells to other cells in local and distant microenvironments. However, the potential of exosomes can be used by engineering them and considering different therapeutic approaches to overcome tumors. Exosomes are a promising drug delivery approach that can help decrease side effects from traditional treatments like radiation and chemotherapy by acting as targeted agents at the tumor site. The present review provides an overview of exosomes and various aspects of the role of exosomes in cancer development, which include these items: exosomes in cancer diagnosis, exosomes and drug delivery, exosomes and drug resistance, exosomal microRNAs and exosomes in tumor microenvironment, etc. Cancer stem cells release exosomes that nurture tumors, promoting unwanted growth and regeneration, and these types of exosomes should be inhibited. Ironically, exosomes from other cells, such as hepatocytes or mesenchymal stem cells (MSCs), are vital for healing organs like the liver and repairing gastric ulcers. Without proper treatment, this healing process can backfire, potentially leading to disease progression or even cancer. What can be found from various studies about the role of exosomes in the field of cancer is that exosomes act like a double-edged sword; on the other hand, natural exosomes in the body may play an important role in the process and progression of cancer, but by engineering exosomes, they can be directed towards target therapy and targeted delivery of drugs to tumor cells. By examining the role and application of exosomes in various mechanisms of cancer, it is possible to help treat this disease more efficiently and quickly in preclinical and clinical research.
Collapse
Affiliation(s)
- Mohsen Safaei
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Seyedeh Somayeh Rajabi
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Mahtab Tirgar
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Najmeh Namdar
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Mahsa Dalfardi
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Farnia Mohammadifar
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Arash Goodarzi
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Ahmad Reza Farmani
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Vahid Ramezani
- Department of Pharmaceutics, School of Pharmacy, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Zahra Abpeikar
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Fasa University of Medical Sciences, Fasa, Iran
| |
Collapse
|
5
|
Qiu H, Liu F, Qiu M, Yang J, Peng X. Monotropein attenuates renal cell carcinoma cell progression and M2 macrophage polarization by weakening NF-κB. Int Urol Nephrol 2025; 57:1785-1795. [PMID: 39776402 DOI: 10.1007/s11255-024-04358-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 12/24/2024] [Indexed: 01/11/2025]
Abstract
PURPOSE The study aimed to investigate the effect and mechanism of monotropein on renal cell carcinoma (RCC). METHODS After monotropein and NF-κB receptor activator (RANKL) treatment, cell proliferation, invasion, and apoptosis were evaluated using CCK-8, Transwell, and flow cytometry. Primary macrophages co-cultured with monotropein-treated RCC cells were analyzed to evaluate macrophage polarization using qRT-PCR, western blot, and ELISA assays by detecting the expression of M2 markers (CD206, CD168) and cytokines (IL-10, TGF-β). Additionally, the therapeutic efficacy of monotropein was examined using an RCC mouse xenograft model. RESULTS Monotropein could inhibit the proliferation, invasion, and M2 macrophage polarization and accelerate the apoptosis of RCC cells. Mechanistically, monotropein suppressed NF-κB pathway activation in RCC cells and reduced the expression of NF-κB downstream targets, including Bcl-2, c-Myc, and MMP9. RANKL could eliminate the effect of monotropein on RCC progression. In primary macrophages co-cultured with monotropein-treated RCC cells, monotropein downregulated M2 polarization markers and cytokines, further supporting its role in modulating the tumor microenvironment. In mouse models, monotropein reduced RCC tumor growth, induced apoptosis, and blocked NF-κB pathway. CONCLUSIONS Monotropein prevents RCC malignant progression and reduces M2 macrophage polarization by suppressing the NF-κB pathway, suggesting that monotropein may serve as a potential therapeutic agent for RCC by targeting both tumor cells and the tumor microenvironment.
Collapse
Affiliation(s)
- Heping Qiu
- Department of Urology, The Second Affiliated Hospital of Nanchang University, No.1, Minde Road, East Lake District, Nanchang, 330006, Jiangxi, China
| | - Fei Liu
- Department of Urology, The Second Affiliated Hospital of Nanchang University, No.1, Minde Road, East Lake District, Nanchang, 330006, Jiangxi, China
| | - Mei Qiu
- Department of Urology, Shanghai Changzheng Hospital, Shanghai, 341099, China
| | - Juliang Yang
- Department of Urology, The Affiliated Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou, 341099, China
| | - Xiang Peng
- Department of Urology, The Second Affiliated Hospital of Nanchang University, No.1, Minde Road, East Lake District, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
6
|
Yang Y, Li S, To KKW, Zhu S, Wang F, Fu L. Tumor-associated macrophages remodel the suppressive tumor immune microenvironment and targeted therapy for immunotherapy. J Exp Clin Cancer Res 2025; 44:145. [PMID: 40380196 DOI: 10.1186/s13046-025-03377-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 03/27/2025] [Indexed: 05/19/2025] Open
Abstract
Despite the significant advances in the development of immune checkpoint inhibitors (ICI), primary and acquired ICI resistance remains the primary impediment to effective cancer immunotherapy. Residing in the tumor microenvironment (TME), tumor-associated macrophages (TAMs) play a pivotal role in tumor progression by regulating diverse signaling pathways. Notably, accumulating evidence has confirmed that TAMs interplay with various cellular components within the TME directly or indirectly to maintain the dynamic balance of the M1/M2 ratio and shape an immunosuppressive TME, consequently conferring immune evasion and immunotherapy tolerance. Detailed investigation of the communication network around TAMs could provide potential molecular targets and optimize ICI therapies. In this review, we systematically summarize the latest advances in understanding the origin and functional plasticity of TAMs, with a focus on the key signaling pathways driving macrophage polarization and the diverse stimuli that regulate this dynamic process. Moreover, we elaborate on the intricate interplay between TAMs and other cellular constituents within the TME, that is driving tumor initiation, progression and immune evasion, exploring novel targets for cancer immunotherapy. We further discuss current challenges and future research directions, emphasizing the need to decode TAM-TME interactions and translate preclinical findings into clinical breakthroughs. In conclusion, while TAM-targeted therapies hold significant promise for enhancing immunotherapy outcomes, addressing key challenges-such as TAM heterogeneity, context-dependent plasticity, and therapeutic resistance-remains critical to achieving optimal clinical efficacy.
Collapse
Affiliation(s)
- Yan Yang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Sijia Li
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Kenneth K W To
- School of Pharmacy, The Chinese University of Hong Kong, Hong Kong, 999077, P.R. China
| | - Shuangli Zhu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Fang Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Liwu Fu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China.
| |
Collapse
|
7
|
Xiang J, Yao L, Wang S, Zhao L, Yu J. Progress of exosomes in regulating tumor metastasis by remodeling the pre-metastatic immune microenvironment. Cell Immunol 2025; 413:104960. [PMID: 40367831 DOI: 10.1016/j.cellimm.2025.104960] [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: 02/21/2025] [Revised: 04/15/2025] [Accepted: 04/24/2025] [Indexed: 05/16/2025]
Abstract
Exosomes play an important role in the metastatic microenvironment, acting as a transmission belt that facilitates intercellular communication. By delivering proteins, nucleic acids, and other substances in the exosomes, they can change the function of the receptor target cells, change the microenvironment of the metastatic site, and promote the colonization of the tumor cells, thus promoting cancer metastasis. The interaction between tumor cells and the surrounding microenvironment is complex, with exosomes serving as key facilitators of crosstalk between the primary tumor microenvironment and the pre-metastasis microenvironment. Despite many current studies to explore exosomes, we still do not have a detailed understanding of the role and mechanism of exosomes in the pre-metastatic immune microenvironment, and there are many challenges in the clinical application of exosomes. In this paper, we summarize the role of exosomes in regulating the pre-metastatic immune microenvironment and its mechanism, focusing on how exosomes regulate the function of immune cells in the pre-metastatic microenvironment to promote tumor metastasis. In addition, the potential application of exosomes in tumor immunotherapy and strategies for targeting exosomes are discussed. This will contribute to the immunotherapy of cancer metastasis and promote the clinical application of exosomes.
Collapse
Affiliation(s)
- Jiangning Xiang
- Cancer Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong an Road, Xi Cheng District, Beijing 100053, China
| | - Lin Yao
- Cancer Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong an Road, Xi Cheng District, Beijing 100053, China
| | - Shan Wang
- Cancer Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong an Road, Xi Cheng District, Beijing 100053, China
| | - Lei Zhao
- Cancer Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong an Road, Xi Cheng District, Beijing 100053, China.
| | - Jing Yu
- Cancer Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong an Road, Xi Cheng District, Beijing 100053, China.
| |
Collapse
|
8
|
Bai X, Guo YR, Zhao ZM, Li XY, Dai DQ, Zhang JK, Li YS, Zhang CD. Macrophage polarization in cancer and beyond: from inflammatory signaling pathways to potential therapeutic strategies. Cancer Lett 2025; 625:217772. [PMID: 40324582 DOI: 10.1016/j.canlet.2025.217772] [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: 02/26/2025] [Revised: 04/30/2025] [Accepted: 05/02/2025] [Indexed: 05/07/2025]
Abstract
Macrophages are innate immune cells distributed throughout the body that play vital roles in organ development, tissue homeostasis, and immune surveillance. Macrophages acquire a binary M1/M2 polarized phenotype through signaling cascades upon sensing different signaling molecules in the environment, thereby playing a core role in a series of immune tasks, rendering precise regulation essential. M1/M2 macrophage phenotypes regulate inflammatory responses, while controlled activation of inflammatory signaling pathways is involved in regulating macrophage polarization. Among the relevant signaling pathways, we focus on the six well-characterized NF-κB, MAPK, JAK-STAT, PI3K/AKT, inflammasome, and cGAS-STING inflammatory pathways, and elucidate their roles and crosstalk in macrophage polarization. Furthermore, the effects of many environmental signals that influence macrophage polarization are investigated by modulating these pathways in vivo and in vitro. We thus detail the physiological and pathophysiological status of these six inflammatory signaling pathways and involvement in regulating macrophage polarization in cancer and beyond, as well as describe potential therapeutic approaches targeting these signaling pathways. In this review, the latest research advances in inflammatory signaling pathways regulating macrophage polarization are reviewed, as targeting these inflammatory signaling pathways provides suitable strategies to intervene in macrophage polarization and various tumor and non-tumor diseases.
Collapse
Affiliation(s)
- Xiao Bai
- Department of Surgical Oncology, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Yun-Ran Guo
- Health Sciences Institute of China Medical University, Shenyang 110122, China
| | - Zhe-Ming Zhao
- Department of Surgical Oncology, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Xin-Yun Li
- Clinical Medicine, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Dong-Qiu Dai
- Department of Surgical Oncology, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China; Cancer Center, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| | - Jia-Kui Zhang
- Department of Surgical Oncology, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| | - Yong-Shuang Li
- Department of Surgical Oncology, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| | - Chun-Dong Zhang
- Department of Surgical Oncology, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China; Central Laboratory, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| |
Collapse
|
9
|
Azar BKY, Vakhshiteh F. The Pre-metastatic Niche: How Cancer Stem Cell-Derived Exosomal MicroRNA Fit into the Puzzle. Stem Cell Rev Rep 2025; 21:1062-1074. [PMID: 40095238 DOI: 10.1007/s12015-025-10866-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/12/2025] [Indexed: 03/19/2025]
Abstract
Cancer metastasis is a complicated biological process that critically affects cancer progression, patient outcomes, and treatment plans. A significant step in metastasis is the formation of a pre-metastatic niche (PMN). A small subset of cells within tumors, known as cancer stem cells (CSCs), possess unique characteristics including, differentiation into different cell types within the tumor, self-renewal, and resistance to conventional therapies, that enable them to initiate tumors and drive metastasis. PMN plays an important role in preparing secondary organs for the arrival and proliferation of CSCs, thereby facilitating metastasis. CSC-derived exosomes are crucial components in the complex interplay between CSCs and the tumor microenvironment. These exosomes function as transporters of various substances that can promote cancer progression, metastasis, and modulation of pre-metastatic environments by delivering microRNA (miRNA, miR) cargo. This review aims to illustrate how exosomal miRNAs (exo-miRs) secreted by CSCs can predispose PMN and promote angiogenesis and metastasis.
Collapse
Affiliation(s)
- Behjat Kheiri Yeghaneh Azar
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Faezeh Vakhshiteh
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran.
| |
Collapse
|
10
|
Liu M, Li TZ, Xu C. The role of tumor-associated fibroblast-derived exosomes in chemotherapy resistance of colorectal cancer and its application prospect. Biochim Biophys Acta Gen Subj 2025; 1869:130796. [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] [MESH Headings] [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 Gastroenterology, The second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, NanChang, China
| | - Congcong Xu
- Department of Cardiovascular Medicine, The second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, NanChang, China.
| |
Collapse
|
11
|
Song G, Zeng C, Li J, Liu J, Zhao J, Liu B, Fan J, Xie H. Exosome-based nanomedicines for digestive system tumors therapy. Nanomedicine (Lond) 2025; 20:1167-1180. [PMID: 40248953 PMCID: PMC12068745 DOI: 10.1080/17435889.2025.2493037] [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: 01/18/2025] [Accepted: 04/10/2025] [Indexed: 04/19/2025] Open
Abstract
Digestive system tumors constitute a major subset of malignancies, consistently ranking among the leading causes of mortality globally. Despite limitations inherent in current therapeutic modalities, recent advancements in targeted therapy and drug delivery systems have led to significant improvements in the efficacy of pharmacotherapy for digestive system tumors. In this context, exosomes - naturally occurring nanoscale vesicles - have emerged as promising drug delivery candidates due to their intrinsic molecular transport capabilities, superior biocompatibility, and targeted recognition of tumor cells. The integration of exosomes into cancer therapeutics represents a novel and potentially transformative approach for treating digestive system tumors, which may drive further progress in this field. This review comprehensively examines the sources, loading mechanisms, and therapeutic efficacy of exosomes in the context of digestive system tumor treatment. Furthermore, it discusses the opportunities and challenges associated with exosomes, offering insights into their future role within the therapeutic armamentarium against digestive tumors.
Collapse
Affiliation(s)
- Ge Song
- Key Laboratory of Cancer Cellular and Molecular Pathology in Hunan Province, Cancer Research Institute of Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Chenlu Zeng
- Key Laboratory of Cancer Cellular and Molecular Pathology in Hunan Province, Cancer Research Institute of Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Junru Li
- Key Laboratory of Cancer Cellular and Molecular Pathology in Hunan Province, Cancer Research Institute of Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Jiajia Liu
- Key Laboratory of Cancer Cellular and Molecular Pathology in Hunan Province, Cancer Research Institute of Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Juanxia Zhao
- Department of Pathology, The Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Bin Liu
- College of Biology, Hunan University, Changsha, Hunan, China
| | - Jialong Fan
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha, China
| | - Hailong Xie
- Key Laboratory of Cancer Cellular and Molecular Pathology in Hunan Province, Cancer Research Institute of Hengyang Medical College, University of South China, Hengyang, Hunan, China
| |
Collapse
|
12
|
Li W, Wei H, Liu J, Zhao Z, Wang F, Qiao L, Yin S, Zhang C, Huo M. Exosomal Biglycan promotes gastric cancer progression via M2 polarization and CXCL10-mediated JAK/STAT1 activation. Cancer Lett 2025; 626:217758. [PMID: 40311912 DOI: 10.1016/j.canlet.2025.217758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 04/25/2025] [Accepted: 04/29/2025] [Indexed: 05/03/2025]
Abstract
Recent advancements in tumor immunotherapy have highlighted the importance of the tumor microenvironment in modulating immune responses against cancer cells. Within the TME, macrophages - particularly the M2 phenotype - serve pivotal regulatory functions through cytokine/chemokine secretion to modulate tumor progression. Elucidating the molecular crosstalk between gastric cancer (GC) cells and tumor-associated macrophages (TAMs) remains imperative for developing targeted therapeutic interventions. In this study, we identified Biglycan (BGN), a small leucine-rich proteoglycan, as a key mediator in GC progression. Exosomal BGN derived from GC cell is delivered to macrophages, where it binds to NONO protein, thereby driving M2 polarization and upregulating CXCL10 expression. Elevated CXCL10 levels activate the JAK/STAT1 signaling pathways, thereby potentiating GC cell proliferation, invasion, and metastatic dissemination. Clinically, elevated BGN expression correlates with advanced tumor stage and poor prognosis in GC patients, positioning it as a promising therapeutic target. Our findings reveal a previously unrecognized mechanism of exosomal BGN-mediated M2 macrophage reprogramming and CXCL10-driven oncogenic signaling in the GC microenvironment. These insights establish a novel therapeutic paradigm for GC management through disruption of tumor-macrophage communication.
Collapse
Affiliation(s)
- Wenchao Li
- Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, 528406, China; The Department of Thyroid and Breast Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510000, China
| | - Hongfa Wei
- Department of General Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, 515063, China
| | - Junjie Liu
- Department of Gastric Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510510, China
| | - Zidan Zhao
- Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, 528406, China
| | - Fuhui Wang
- Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, 528406, China
| | - Liang Qiao
- Storr Liver Centre, Westmead Institute for Medical Research, University of Sydney at Westmead Hospital, Westmead, NSW, 2145, Australia.
| | - Songcheng Yin
- Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, 528406, China; Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, 528406, China.
| | - Changhua Zhang
- Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, 528406, China; Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, 528406, China.
| | - Mingyu Huo
- Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, 528406, China; Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, 528406, China.
| |
Collapse
|
13
|
Barreto Garcia V, Gasparotto LHS, de Araujo AA, Leitão RFC, Brito GAC, Vilar NF, Lima Oliveira E, Guedes PMM, de Araújo Júnior RF. Gold Nanoparticles (AuNPs) Coadministered with a β-Blocker Prevent Liver Fibrosis Caused by Ethanol and Methamphetamine in Rats by Downregulating the Expression of M2 Macrophages. ACS OMEGA 2025; 10:14924-14939. [PMID: 40290979 PMCID: PMC12019731 DOI: 10.1021/acsomega.4c10118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 03/21/2025] [Accepted: 04/02/2025] [Indexed: 04/30/2025]
Abstract
Simultaneous abuse of ethanol and methamphetamine (METH) causes severe liver damage through oxidative stress and inflammation. This study evaluated the antifibrotic effects of gold nanoparticles (AuNPs) coadministered with the β-blocker carvedilol (CARV) against liver damage in rats. Male Wistar rats received 30% ethanol (7 g/kg) daily for 28 days, with METH (10 mg/kg) administered on the 22nd and 28th days. Liver damage was assessed using serum hepatic enzymes, glutathione (GSH) levels, malondialdehyde (MDA) formation, myeloperoxidase (MPO) inhibition, and histopathological analysis, including H&E, Picrosirius Red staining, immunofluorescence, and transmission electron microscopy. Cytokine levels were measured in liver tissue samples. In vitro, RAW 264.7 macrophages were induced to polarize into M1 and M2 phenotypes and cocultured with AuNPs + CARV-treated 3T3 cells, analyzed by rtPCR. AuNPs + CARV effectively protected the liver by modulating interactions between hepatic stellate cells (HSCs) and Kupffer cells, promoting an antifibrotic immune response driven by M1 macrophages. This was indicated by downregulation of profibrotic M2 macrophages and upregulation of M1 macrophages, shown by an increased CD86/CD163 ratio and reduced levels of IL-1β, TNF-α, TGFβ, AKT, and PI3K., pointing an attenuated liver inflammation. These results suggest that AuNPs combined with CARV could potentially serve as a therapy for alcohol and METH-induced liver fibrosis by targeting M2 macrophages.
Collapse
Affiliation(s)
- Vinícius Barreto Garcia
- Inflammation
and Cancer Research Laboratory, Department of Morphology, Federal University of Rio Grande do Norte (UFRN), Natal 59078-970, RN, Brazil
| | - Luiz H. S. Gasparotto
- Institute
of Chemistry, Federal University of Mato
Grosso (UFMT), Cuiaba 78060-900, MT, Brazil
| | - Aurigena A. de Araujo
- Department
of Pharmacology, Federal University of Rio
Grande do Norte (UFRN), Natal 59078-970, RN, Brazil
| | - Renata F. C. Leitão
- Department
of Morphology, Postgraduate Program in Morphology, Federal University of Ceará (UFC), Fortaleza 60355-636, CE, Brazil
| | - Gerly A. C. Brito
- Department
of Morphology, Postgraduate Program in Morphology, Federal University of Ceará (UFC), Fortaleza 60355-636, CE, Brazil
| | - Natalia Feitosa Vilar
- Inflammation
and Cancer Research Laboratory, Department of Morphology, Federal University of Rio Grande do Norte (UFRN), Natal 59078-970, RN, Brazil
| | - Emily Lima Oliveira
- Inflammation
and Cancer Research Laboratory, Department of Morphology, Federal University of Rio Grande do Norte (UFRN), Natal 59078-970, RN, Brazil
| | - Paulo M. M. Guedes
- Department
of Microbiology and Parasitology, Federal
University of Rio Grande do Norte (UFRN), Natal 59078-970, RN, Brazil
| | - Raimundo F. de Araújo Júnior
- Inflammation
and Cancer Research Laboratory, Department of Morphology, Federal University of Rio Grande do Norte (UFRN), Natal 59078-970, RN, Brazil
| |
Collapse
|
14
|
Jia H, Li P, Li M, Liu N, Dong J, Qu Q, Cao Z. Trans-Kingdom RNA Dialogues: miRNA and milRNA Networks as Biotechnological Tools for Sustainable Crop Defense and Pathogen Control. PLANTS (BASEL, SWITZERLAND) 2025; 14:1250. [PMID: 40284138 PMCID: PMC12030539 DOI: 10.3390/plants14081250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2025] [Revised: 04/15/2025] [Accepted: 04/18/2025] [Indexed: 04/29/2025]
Abstract
MicroRNAs (miRNAs) are a class of non-coding RNAs approximately 20-24 nucleotides in length, which play a crucial role during gene regulation in plant-pathogen interaction. They negatively regulate the expression of target genes, primarily at the transcriptional or post-transcriptional level, through complementary base pairing with target gene sequences. Recent studies reveal that during pathogen infection, miRNAs produced by plants and miRNA-like RNAs (milRNAs) produced by fungi can regulate the expression of endogenous genes in their respective organisms and undergo trans-kingdom transfer. They can thereby negatively regulate the expression of target genes in recipient cells. These findings provide novel perspectives for deepening our understanding of the regulatory mechanisms underlying plant-pathogen interactions. Here, we summarize and discuss the roles of miRNAs and milRNAs in mediating plant-pathogen interactions via multiple pathways, providing new insights into the functions of these RNAs and their modes of action. Collectively, these insights lay a theoretical foundation for the targeted management of crop diseases.
Collapse
Affiliation(s)
- Hui Jia
- State Key Laboratory of North China Crop Improvement and Regulation, Hebei Agricultural University, Baoding 071000, China; (H.J.); (P.L.); (N.L.); (J.D.)
| | - Pan Li
- State Key Laboratory of North China Crop Improvement and Regulation, Hebei Agricultural University, Baoding 071000, China; (H.J.); (P.L.); (N.L.); (J.D.)
| | - Minye Li
- College of Life Sciences, Fujian Normal University, Fuzhou 350117, China;
| | - Ning Liu
- State Key Laboratory of North China Crop Improvement and Regulation, Hebei Agricultural University, Baoding 071000, China; (H.J.); (P.L.); (N.L.); (J.D.)
| | - Jingao Dong
- State Key Laboratory of North China Crop Improvement and Regulation, Hebei Agricultural University, Baoding 071000, China; (H.J.); (P.L.); (N.L.); (J.D.)
| | - Qing Qu
- College of Agriculture and Forestry, Hebei North University, Zhangjiakou 075000, China
| | - Zhiyan Cao
- State Key Laboratory of North China Crop Improvement and Regulation, Hebei Agricultural University, Baoding 071000, China; (H.J.); (P.L.); (N.L.); (J.D.)
| |
Collapse
|
15
|
Saeed BI, Uthirapathy S, Kubaev A, Ganesan S, Shankhyan A, Gupta S, Joshi KK, Kariem M, Jasim AS, Ahmed JK. Ferroptosis as a key player in the pathogenesis and intervention therapy in liver injury: focusing on drug-induced hepatotoxicity. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04115-w. [PMID: 40244448 DOI: 10.1007/s00210-025-04115-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Accepted: 03/27/2025] [Indexed: 04/18/2025]
Abstract
Globally, drug-induced hepatotoxicity or drug-induced liver injury (DILI) is a serious clinical concern. Knowing the processes and patterns of cell death is essential for finding new therapeutic targets since there are not many alternatives to therapy for severe liver lesions. Excessive lipid peroxidation is a hallmark of ferroptosis, an iron-reliant non-apoptotic cell death linked to various liver pathologies. When iron is pathogenic, concomitant inflammation may exacerbate iron-mediated liver injury, and the hepatocyte necrosis that results is a key element in the fibrogenic response. The idea that dysregulated metabolic pathways and compromised iron homeostasis contribute to the development of liver injury by ferroptosis is being supported by new data. Various ferroptosis-linked genes and pathways have been linked to liver injury, although the molecular processes behind ferroptosis's pathogenicity are not well known. Here, we delve into the features of ferroptosis, the processes governing ferroptosis, and our current knowledge of iron metabolism. We also provide an overview of ferroptosis's involvement in the pathophysiology of liver injury, particularly DILI. Lastly, the therapeutic possibilities of ferroptosis targeting for liver injury management have been provided. Natural products, nanoparticles (NPs), mesenchymal stem cell (MSC), and their exosomes have attracted increasing attention among such therapeutics.
Collapse
Affiliation(s)
- Bahaa Ibrahim Saeed
- Medical Laboratory Techniques Department, College of Health and Medical Technology, University of Al-Maarif, Anbar, Iraq
| | - Subasini Uthirapathy
- Pharmacy Department, Tishk International University, Erbil, Kurdistan Region, Iraq
| | - Aziz Kubaev
- Department of Maxillofacial Surgery, Samarkand State Medical University, 18 Amir Temur Street, 140100, Samarkand, Uzbekistan.
| | - Subbulakshmi Ganesan
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Aman Shankhyan
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Sofia Gupta
- Department of Chemistry, Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, Mohali, 140307, Punjab, India
| | - Kamal Kant Joshi
- Department of Allied Science, Graphic Era Hill University, Dehradun, India
- Graphic Era Deemed to be University, Dehradun, Uttarakhand, India
| | - Muthena Kariem
- Department of Medical Analysis, Medical Laboratory Technique College, the Islamic University, Najaf, Iraq
| | - Ahmed Salman Jasim
- Radiology Techniques Department College of Health and Medical Techniques, Al-Mustaqbal University, 51001, Babylon, Iraq
| | - Jawad Kadhim Ahmed
- Department of Medical Laboratories Technology, AL-Nisour University College, Baghdad, Iraq
| |
Collapse
|
16
|
Adamecz DI, Veres É, Papp C, Árva H, Rónavári A, Marton A, Vizler C, Gácser A, Kónya Z, Igaz N, Kiricsi M. Gold and Silver Nanoparticles Efficiently Modulate the Crosstalk Between Macrophages and Cancer Cells. Int J Nanomedicine 2025; 20:4777-4802. [PMID: 40255669 PMCID: PMC12009049 DOI: 10.2147/ijn.s508171] [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: 12/10/2024] [Accepted: 04/02/2025] [Indexed: 04/22/2025] Open
Abstract
Background Macrophages, polarized into pro-inflammatory M1 or anti-inflammatory M2 states, are essential cellular elements of innate immunity. In the tumor microenvironment, owing to a paracrine manipulative program by cancerous cells, tumor-associated macrophages (TAMs) evolve, which can shift between M1-like and M2-like phenotypes. Since it is fairly unknown how the promising anticancer agents, silver (AgNPs) and gold nanoparticles (AuNPs) affect the bidirectional communication and reprogramming in the tumor stroma, we examined the behavior, the tumor-supporting functions, and the expression of polarization and functional marker genes of TAMs to reveal how these are modulated upon interaction with nanoparticle-exposed cancer cells. Methods We established co-cultures of murine immortalized J774 or primary bone marrow-derived macrophages with 4T1 breast cancer cells treated with AuNPs or AgNPs or with none of the nanoparticles. We assessed the expression of macrophage polarization and functional markers using RT-qPCR and Proteome Profiler Array and evaluated macrophage migration and matrix metalloproteinase activity by specific assays. Results Protein and mRNA levels of most examined factors - except tumor necrosis factor-alpha - such as C-C-motif chemokine ligands 2 and 22, interleukin-23, inducible nitric oxide synthase, cyclooxygenase-2, the macrophage mannose receptor CD206, transforming growth factor-beta, and chitinase-like-3 protein decreased, and the expression of polarization markers revealed a shift towards M1-like phenotype in macrophages co-cultured with AgNP- or AuNP-treated 4T1 cells. Both nanoparticle treatments reduced the levels and activity of cell migration-related factors, such as C-C motif chemokine ligand 3, matrix metalloproteinases, and suppressed macrophage migration. Conclusion Both AuNPs and AgNPs showed a remarkable ability to influence macrophage-cancer cell communication, suppressed indirectly M2-like TAM polarization, and perturbed the migration behavior of TAMs that is critical for tumor invasion, indicating modulated immunological functions and debilitated cancer-promoting capabilities of TAMs in this microenvironment.
Collapse
Affiliation(s)
- Dóra Izabella Adamecz
- Department of Biochemistry and Molecular Biology, University of Szeged, Szeged, Hungary
- Doctoral School of Biology, University of Szeged, Szeged, Hungary
| | - Éva Veres
- Doctoral School of Biology, University of Szeged, Szeged, Hungary
- Department of Biotechnology and Microbiology, University of Szeged, Szeged, Hungary
- HCEMM-SZTE Pathogen Fungi Research Group, University of Szeged, Szeged, Hungary
| | - Csaba Papp
- Department of Biotechnology and Microbiology, University of Szeged, Szeged, Hungary
- HCEMM-SZTE Pathogen Fungi Research Group, University of Szeged, Szeged, Hungary
| | - Hédi Árva
- Department of Biochemistry and Molecular Biology, University of Szeged, Szeged, Hungary
- Doctoral School of Biology, University of Szeged, Szeged, Hungary
| | - Andrea Rónavári
- Department of Applied and Environmental Chemistry, University of Szeged, Szeged, Hungary
| | - Annamária Marton
- Laboratory of Tumor Immunology and Pharmacology, Centre of Excellence of the European Union, HUN-REN Biological Research Centre, Szeged, Hungary
| | - Csaba Vizler
- Laboratory of Tumor Immunology and Pharmacology, Centre of Excellence of the European Union, HUN-REN Biological Research Centre, Szeged, Hungary
| | - Attila Gácser
- Department of Biotechnology and Microbiology, University of Szeged, Szeged, Hungary
- HCEMM-SZTE Pathogen Fungi Research Group, University of Szeged, Szeged, Hungary
| | - Zoltán Kónya
- Department of Applied and Environmental Chemistry, University of Szeged, Szeged, Hungary
| | - Nóra Igaz
- Department of Biochemistry and Molecular Biology, University of Szeged, Szeged, Hungary
| | - Mónika Kiricsi
- Department of Biochemistry and Molecular Biology, University of Szeged, Szeged, Hungary
| |
Collapse
|
17
|
Bai T, Li P, Liu Y, Cai B, Li G, Wang W, Yan R, Zheng X, Du S. Knockdown of miR-411-3p induces M2 macrophage polarization and promotes colorectal cancer progression by regulation of MMP7. Eur J Histochem 2025; 69:4178. [PMID: 40322788 PMCID: PMC12086358 DOI: 10.4081/ejh.2025.4178] [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: 12/26/2024] [Accepted: 04/13/2025] [Indexed: 05/21/2025] Open
Abstract
Colorectal cancer (CRC) is prone to metastasis, leading to a poor prognosis. miR-411-3p exhibits a tumor-suppressive function in CRC, but its exact mechanism is unclear. The malignant biological properties of CRC cells were detected by Carboxyfluorescein diacetate succinimidyl ester (CFSE) staining, scratch-wound and transwell assay. Levels of markers associated with macrophage polarization were evaluated by flow cytometry and ELISA kits. Bioinformatics analysis to screen whether the downstream target mRNA of miR-411-3p is matrix metalloproteinase 7 (MMP7), and Dual-Luciferase reporter assay verified the targeting relationship between the two. qRT-PCR tested miR-411-3p and MMP7 levels. MMP7 level was quantified by Western blot. Additionally, a nude mouse subcutaneous graft tumor model was constructed, Ki-67 expression was detected by immunohistochemistry, and the impact of miR-411-3p/MMP7 on the polarization of M2 macrophages was explored. miR-411-3p expression is downregulated in CRC. Knockdown of miR-411-3p elevated the amount of CFSE-positive, migrating, and invading cells, decreased apoptosis, and elevated the levels of M2 macrophage polarization markers. After overexpression of miR-411-3p, all of the above metrics were reversed in CRC cells. miR-411-3p targeted negative regulation of MMP7 expression, and MMP7 overexpression further enhanced the promotional effect of knockdown of miR-411-3p on the malignant progression of CRC and M2 macrophage polarization. Furthermore, knockdown of miR-411-3p upregulated the MMP7 level, elevated Ki-67-positive cells count, and induced M2 macrophage polarization in vivo. Knockdown of miR-411-3p upregulates MMP7 and induces M2 macrophage polarization, which in turn promotes malignant biological progression of CRC.
Collapse
Affiliation(s)
- Tianliang Bai
- Department of Gastrointestinal Surgery, Affiliated Hospital of Hebei University, Baoding, Hebei Province
| | - Ping Li
- Department of Gastrointestinal Surgery, Affiliated Hospital of Hebei University, Baoding, Hebei Province
| | - Yabin Liu
- Department of General Surgery, Fourth Hospital of Hebei Medical University, Hebei Province
| | - Bindan Cai
- Department of Neurology, Zhuozhou City Hospital, Hebei Province, China
| | - Gang Li
- Department of Gastrointestinal Surgery, Affiliated Hospital of Hebei University, Baoding, Hebei Province
| | - Wenbin Wang
- Department of Gastrointestinal Surgery, Affiliated Hospital of Hebei University, Baoding, Hebei Province
| | - Rui Yan
- Department of Gastrointestinal Surgery, Affiliated Hospital of Hebei University, Baoding, Hebei Province
| | - Xiangkui Zheng
- Department of Gastrointestinal Surgery, Affiliated Hospital of Hebei University, Baoding, Hebei Province
| | - Shangkun Du
- Department of Gastrointestinal Surgery, Affiliated Hospital of Hebei University, Baoding, Hebei Province
| |
Collapse
|
18
|
Long K, Kui X, Zeng Q, Dong W. Cancer cell-derived exosomal miR-34a inhibits the malignant progression of pancreatic adenocarcinoma cells by restraining the M2 polarization of macrophages. Eur J Histochem 2025; 69:4176. [PMID: 40244037 PMCID: PMC12051414 DOI: 10.4081/ejh.2025.4176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 04/02/2025] [Indexed: 04/18/2025] Open
Abstract
This study aimed to investigate the crosstalk mechanism between pancreatic cancer (PAC) cells and M2 tumor-associated macrophages induced by tumor-derived exosomal miR-34a. MicroRNA and mRNA expression levels were detected using RT-qPCR. Cell Counting Kit-8, wound-healing, transwell assays and flow cytometry were respectively employed to assess cell proliferation, migration, invasion and apoptosis. Enzyme-linked immunosorbent assay was utilized to determine cytokine secretion. Transmission electron microscopy and nanoparticle tracking analyses were performed to detect the exosome morphology and particle size. Phagocytosis of exosomes by macrophages was verified by PKH26 labeling. The effects of exosome-treated macrophages on the epithelial-mesenchymal transition, invasion, and migration of PANC-1 cells were investigated using coculture experiments. The identification of miR-34a's potential targets were determined with TargetScan and validated by a dual-luciferase reporter assay. miR-34a was expressed at low levels in PAC tissues, cells, and exosomes. The overexpression of miR-34a restrains the malignant progression of PANC-1 cells. After miR-34a-overexpressed PANC-1-derived exosomes were phagocytosed by macrophages, the process of M2 polarization in macrophages was obstructed, leading to the suppression of epithelial-mesenchymal transition, migration, and invasion of the cocultured PANC-1 cells. Suppressor of cytokine signaling 3 is a direct target of miR-34a. MiR-34a negatively modulates the suppressor of cytokine signaling 3 to prevent the M2 polarization of macrophages by engaging the Janus kinase/signal transducers and activators of the transcription pathway and influencing the malignancy of PAC cells. miR-34a in cancer cell-derived exosomes inhibits the malignant progression of pancreatic cancer cells by restraining M2 polarization of macrophages.
Collapse
Affiliation(s)
- Kui Long
- Department of Three Wards of Hepatobiliary Surgery
| | - Xiang Kui
- Department of Pathology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Qingbin Zeng
- Department of Three Wards of Hepatobiliary Surgery
| | - Wenzhi Dong
- Department of Three Wards of Hepatobiliary Surgery
| |
Collapse
|
19
|
Pan C, Zhou Z, Cao J, Zhang L, Cheng T, Li H, Jiang Z, Huang D, Zeng D, Luo Y, Wu J. MACC1 is a potential prognostic biomarker for cancer immunotherapy in lung adenocarcinoma. Carcinogenesis 2025; 46:bgaf015. [PMID: 40117327 DOI: 10.1093/carcin/bgaf015] [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: 12/29/2024] [Revised: 03/02/2025] [Accepted: 03/15/2025] [Indexed: 03/23/2025] Open
Abstract
Our team previously reported that MACC1 levels are closely related to a variety of tumors and the efficacy of immune checkpoint blockade (ICB) therapy. However, the predictive value of MACC1 levels for lung adenocarcinoma (LUAD) immunotherapy has not been studied. This study aimed to investigate the predictive effect of the oncogene MACC1 on ICB reactivity in patients with LUAD. First, the expression patterns and clinical features of MACC1 in The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases were comprehensively evaluated using R packages. We subsequently assessed the correlations between MACC1 and immunological characteristics in the LUAD tumor microenvironment (TME) using the CIBERSORT algorithm. The results revealed that MACC1 overexpression was significantly correlated with 3 immune checkpoints, 14 tumor-infiltrating immune cells (TIICs), 9 immunomodulators, 5 anticancer immune process activities, and 3 effector genes of TIICs in LUAD. Additionally, on the basis of the prognostic genes from LASSO analysis, we developed the MACC1-related Risk Score (MRRS), which can accurately predict the prognosis and response to cancer immunotherapy in LUAD patients (HR = 3.50, AUC at 1, 2, and 3 years = 0.737, 0.744, and 0.724, respectively). Finally, in vivo experiments revealed that the combination of MACC1 silencing and PD-L1 inhibitors significantly inhibits tumor progression. These findings increase our understanding of MACC1 as a potential prognostic biomarker and potential therapeutic target for cancer immunotherapy. The MRRS may play a critical role in predicting the response of LUAD patients to ICB therapy.
Collapse
Affiliation(s)
- Changqie Pan
- Thoracic Medicine Department 1, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, No. 283, Tongzipo Road, Yuelu District, Changsha, Hunan 410013, China
| | - Zhiyuan Zhou
- Department of Oncology, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Avenue North, Baiyun District, Guangzhou, Guangdong 510515, China
| | - Jun Cao
- Thoracic Medicine Department 1, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, No. 283, Tongzipo Road, Yuelu District, Changsha, Hunan 410013, China
| | - Lemeng Zhang
- Thoracic Medicine Department 1, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, No. 283, Tongzipo Road, Yuelu District, Changsha, Hunan 410013, China
| | - Tianli Cheng
- Thoracic Medicine Department 1, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, No. 283, Tongzipo Road, Yuelu District, Changsha, Hunan 410013, China
| | - Haitao Li
- Thoracic Medicine Department 1, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, No. 283, Tongzipo Road, Yuelu District, Changsha, Hunan 410013, China
| | - Zhou Jiang
- Thoracic Medicine Department 1, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, No. 283, Tongzipo Road, Yuelu District, Changsha, Hunan 410013, China
| | - Danhui Huang
- Department of Respiratory and Critical Care Medicine, Chronic Airways Diseases Laboratory, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Avenue North, Baiyun District, Guangzhou, Guangdong 510515, China
| | - Dongqiang Zeng
- Department of Oncology, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Avenue North, Baiyun District, Guangzhou, Guangdong 510515, China
| | - Yongzhong Luo
- Thoracic Medicine Department 1, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, No. 283, Tongzipo Road, Yuelu District, Changsha, Hunan 410013, China
| | - Jianhua Wu
- Department of Oncology, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Avenue North, Baiyun District, Guangzhou, Guangdong 510515, China
| |
Collapse
|
20
|
Wei W, Li J, Huang J, Jiang Q, Lin C, Hu R, Wei J, Li Q, Xu G, Chang Z. Exosomal miR‑3681‑3p from M2‑polarized macrophages confers cisplatin resistance to gastric cancer cells by targeting MLH1. Mol Med Rep 2025; 31:94. [PMID: 39981936 PMCID: PMC11851060 DOI: 10.3892/mmr.2025.13459] [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: 08/12/2024] [Accepted: 01/07/2025] [Indexed: 02/22/2025] Open
Abstract
Cisplatin (DDP) is a key chemotherapeutic agent in the treatment of gastric cancer; however, its efficacy is often limited by chemoresistance, a notable challenge in clinical oncology. The present study aimed to investigate the influence of exosomes derived from M2‑polarized macrophages, which promote this resistance, on the response of gastric cancer cells to DDP, examining both the effects and the underlying mechanisms. M2 macrophages, differentiated from mouse bone marrow cells with interleukin (IL)‑13 and IL‑4, were identified using immunofluorescence staining for CD206 and CD163. Exosomes derived from these macrophages were characterized using transmission electron microscopy and protein markers, including calnexin, tumor susceptibility gene 101 and CD9. The role of exosomal microRNA (miR)‑3681‑3p in DDP resistance was assessed using Cell Counting Kit‑8 and apoptosis assays, while a luciferase reporter assay was used to elucidate the interaction between miR‑3681‑3p and MutL protein homolog 1 (MLH1). Co‑culturing gastric cancer cells with M2 macrophages enhanced DDP resistance, an effect amplified by exosomes from M2 macrophages enriched with miR‑3681‑3p. This microRNA directly targeted and reduced MLH1 protein expression. Overexpression of miR‑3681‑3p through mimic transfection, along with MLH1 silencing by small interfering RNA transfection, significantly increased DDP resistance, as evidenced by elevated IC50 values in AGS cells. By contrast, the overexpression of MLH1 effectively reversed the drug resistance of AGS cells to DDP caused by miR‑3681‑3p mimic transfection, as evidenced by a decrease in the IC50 value. In conclusion, exosomal miR‑3681‑3p from M2 macrophages may have a key role in conferring DDP resistance to gastric cancer by suppressing MLH1, offering a new therapeutic target for overcoming chemoresistance.
Collapse
Affiliation(s)
- Wujun Wei
- Center for Clinical Laboratory Diagnosis and Research, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi 533000, P.R. China
- Clinic Medicine Research Center of Hepatobiliary Diseases, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi 533000, P.R. China
- Key Laboratory of Research on Clinical Molecular Diagnosis for High Incidence Diseases in Western Guangxi, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi 533000, P.R. China
- Baise Key Laboratory for Research and Development on Clinical Molecular Diagnosis for High-Incidence Diseases, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi 533000, P.R. China
| | - Jiaxing Li
- Department of Pharmacy, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi 533000, P.R. China
| | - Jingjing Huang
- Department of Health Care, Baise Maternity and Child Health Center, Baise, Guangxi 533000, P.R. China
| | - Qi Jiang
- Department of Gastroenterology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi 533000, P.R. China
| | - Cheng Lin
- Department of Oncology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi 533000, P.R. China
| | - Rentong Hu
- Center for Clinical Laboratory Diagnosis and Research, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi 533000, P.R. China
| | - Jiazhu Wei
- Center for Clinical Laboratory Diagnosis and Research, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi 533000, P.R. China
| | - Qiao Li
- Department of Neurology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi 533000, P.R. China
| | - Guidan Xu
- Center for Clinical Laboratory Diagnosis and Research, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi 533000, P.R. China
| | - Zhengyi Chang
- Center for Clinical Laboratory Diagnosis and Research, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi 533000, P.R. China
| |
Collapse
|
21
|
Liu S. MiR-374a/b-5p Suppresses Cell Growth in Papillary Thyroid Carcinoma Through Blocking Exosomal ANXA1-Induced Macrophage M2 Polarization. Biochem Genet 2025; 63:1258-1274. [PMID: 38536567 DOI: 10.1007/s10528-024-10747-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 02/16/2024] [Indexed: 03/23/2025]
Abstract
Papillary thyroid carcinoma (PTC), comprising 85% of all thyroid cancers, is an epithelial malignancy. The potential for malignant transformation in normal cells by thyroid cancer cells via exosomal Annexin A1 (ANXA1) delivery is investigated in this study. Our aim is to determine the impact of PTC cells on macrophage polarization through exosomal ANXA1 secretion and its implications for tumor progression. Exosomes in PTC cells were examined using transmission electron microscopy, exosome labeling, and nanoparticle tracking analysis. Real-time quantitative polymerase chain reaction was employed to quantify gene expression levels. Protein levels were determined through Western blot analysis. The interplay between genes was assessed using luciferase reporter and RNA pull-down assays. Functional experiments were conducted to investigate PTC cell proliferation and apoptosis. Our findings reveal that ANXA1 promotes PTC cell proliferation and inhibits apoptosis. Exosomes derived from PTC cells were found to promote macrophage M2 polarization. ANXA1 stimulates M2 polarization through the activation of the PI3K/AKT pathway. MicroRNA-374a-5p (miR-374a-5p) and microRNA-374b-5p (miR-374b-5p) were identified as inhibitors of ANXA1 expression and PI3K/AKT pathway activity, thereby inhibiting macrophage M2 polarization. Furthermore, miR-374a-5p and miR-374b-5p were observed to suppress PTC cell proliferation through their regulatory action on ANXA1. Our study suggests that miR-374a/b-5p inhibits PTC cell growth by blocking the macrophage M2 polarization induced by exosomal ANXA1.
Collapse
Affiliation(s)
- Sanbao Liu
- The Second Affiliated Hospital of Wannan Medical College, No.10 Kangfu Road, Wuhu, 241001, Anhui, China.
| |
Collapse
|
22
|
Hu G, Cai P, Li J, Yu L, Zhao B, Chen G. Serum exosomal miR-454-3p contributes to malignant progression of lung cancer by inhibiting HHEX. Mol Cell Probes 2025; 80:102019. [PMID: 39929349 DOI: 10.1016/j.mcp.2025.102019] [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: 07/26/2024] [Revised: 02/08/2025] [Accepted: 02/08/2025] [Indexed: 02/17/2025]
Abstract
BACKGROUND Lung cancer is a common cancer. Exosomes are emerging mediators of intercellular communication, and miRNAs serve a crucial position in cancer progression. This project intends to discover whether exosomal miR-454-3p affects tumor progression and its underlying mechanisms. METHODS Exosomes were isolated utilizing ultracentrifugation. The exosomal biomarkers level was monitored by western blot (WB). The miR-454-3p levels were assessed by quantitative reverse transcription polymerase chain reaction (qRT-PCR), and HHEX expression were detected by qRT-PCR and WB. Cell growth and metastasis were detected through CCK-8, colony formation assay and transwell. Meanwhile, the dual luciferase reporter system and immunoprecipitation (RIP) assay was applied to clarify the interactions between miR-454-3p and HHEX. RESULTS We successfully isolated serum exosomes from NSCLC patients. Then, our team discovered that miR-454-3p was elevated in serum-derived exosomes from NSCLC patients. Functional analysis disclosed that exosomes accelerated NSCLC cell proliferation and metastasis. Silencing of exosomal miR-454-3p hindered NSCLC cell proliferation and metastasis. Subsequently, the starbase database declared that miR-454-3p was interacted with HHEX. HHEX overexpression reversed the promotion of NSCLC cell proliferation and metastasis by exosomal miR-454-3p. CONCLUSIONS Exosomal miR-454-3p enhanced the progression of NSCLC cells through HHEX. miR-454-3p may be a therapeutic target for NSCLC.
Collapse
Affiliation(s)
- Gangqin Hu
- Medical Oncology, Jingmen People's Hospital, Jingmen, 448000, Hubei, China
| | - Peng Cai
- Medical Oncology, Jingmen People's Hospital, Jingmen, 448000, Hubei, China
| | - Jingjing Li
- Medical Oncology, Jingmen People's Hospital, Jingmen, 448000, Hubei, China
| | - Liuyang Yu
- Medical Oncology, Jingmen People's Hospital, Jingmen, 448000, Hubei, China
| | - Bolin Zhao
- Medical Oncology, Jingmen People's Hospital, Jingmen, 448000, Hubei, China
| | - Guiming Chen
- Medical Oncology, Jingmen People's Hospital, Jingmen, 448000, Hubei, China.
| |
Collapse
|
23
|
Barjasteh AH, Jaseb Mazhar AleKassar R, Al-Asady AM, Latifi H, Avan A, Khazaei M, Ryzhikov M, Hassanian SM. Therapeutic Potentials of MiRNA for Colorectal Cancer Liver Metastasis Treatment: A Narrative Review. IRANIAN JOURNAL OF MEDICAL SCIENCES 2025; 50:202-219. [PMID: 40255223 PMCID: PMC12008659 DOI: 10.30476/ijms.2024.102910.3622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 11/01/2024] [Accepted: 11/26/2024] [Indexed: 04/22/2025]
Abstract
Colorectal cancer (CRC) ranks among the most prevalent cancers worldwide and is the fourth leading cause of cancer-related deaths. Metastasis poses a significant obstacle in CRC treatment, as distant metastasis, particularly to the liver, remains the primary cause of mortality. Colorectal liver metastasis (CRLM) occurs frequently due to the liver's direct vascular connection to the colorectal region via the portal vein. Standard treatment approaches for CRLM are limited; only a few patients qualify for surgical intervention, resulting in a persistently low survival rate. Additionally, resistance to chemotherapy is common, emphasizing the need for more effective targeted therapies. Emerging evidence highlights the pivotal role of microRNAs (miRNAs) in modulating critical pathways associated with CRLM, including tumor invasion, epithelial-mesenchymal transition, and angiogenesis. MiRNAs exhibit dual functions as tumor suppressors and oncogenes by targeting multiple genes, thus playing a complex role in both the initiation and progression of metastasis. The regulatory mechanisms of miRNAs could help to identify novel biomarkers for early diagnosis and prognosis of CRLM, as well as promising therapeutic targets to overcome chemoresistance. Despite numerous studies on miRNA involvement in CRC metastasis, dedicated reviews focusing on miRNAs and CRLM remain scarce. This review aims to approach targeted therapies by examining the current understanding of miRNA involvement in CRLM and exploring their potential as diagnostic, prognostic, and therapeutic agents. Through an integrative approach, we aim to provide insights that could transform CRLM management and improve patient outcomes.
Collapse
Affiliation(s)
- Amir Hossein Barjasteh
- Metabolic Syndrome Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Rawa Jaseb Mazhar AleKassar
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Abdulridha Mohammed Al-Asady
- Department of Medical Sciences, Faculty of Nursing, Warith Al-Anbiyaa University, Iraq
- Department of Medical Sciences, Faculty of Dentistry, University of Kerbala, Iraq
- Department of Pharmacology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hanieh Latifi
- Metabolic Syndrome Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Faculty of Health, School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, QLD 4059, Australia
| | - Majid Khazaei
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Seyed Mahdi Hassanian
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
24
|
Han Y, Zhou Z, Li R, Wang H. Tumor-Derived Exosomal circ_0020095 Promotes Colon Cancer Cell Proliferation and Metastasis by Inhibiting M1 Macrophage Polarization. J Biochem Mol Toxicol 2025; 39:e70225. [PMID: 40165503 DOI: 10.1002/jbt.70225] [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: 06/12/2024] [Revised: 01/20/2025] [Accepted: 03/03/2025] [Indexed: 04/02/2025]
Abstract
Tumor-associated macrophages (TAM) have been shown to regulate colon cancer (CC) progression. However, it is not clear whether tumor-derived exosomal circular RNA (circRNA) regulates TAM to influence CC progression. The expression levels of circ_0020095, M1 macrophage markers, M2 macrophage markers, and interleukin-1 receptor-associated kinase 1 (IRAK1) were determined by qRT-PCR. Cell proliferation, migration and invasion were examined by EdU assay, wound healing assay and transwell assay. Exosomes derived from CC cells were used to treat M0 macrophages. M1 macrophage surface marker CD86 was detected by flow cytometry, and protein expression was examined by western blot. Then, the medium of exosome-treated M0 macrophages was used to culture CC cells to determine CC cell functions. RNA pull-down assay, RIP assay and dual-luciferase reporter assay were performed to validate interaction. Circ_0020095 had elevated expression in CC tissues and cells, and its knockdown repressed CC cell proliferation and metastasis. M0 macrophages could take by CC cell-derived exosomes to regulate circ_0020095 expression. Exosomal circ_0020095 restrained M1 macrophage polarization and increased M2 macrophage polarization to enhance CC cell progression. Besides, IRAK1 silencing could promote CC cell proliferation and metastasis by inhibiting M1 macrophage polarization, and its overexpression also abolished the effect of exosomal circ_0020095. Mechanistically, circ_0020095 could competitively bind to IGF2BP1 and then reduced the binding ability of IGF2BP1 and IRAK1 3'UTR. Tumor-derived exosomal circ_0020095 promoted CC cell progression via inhibiting M1 macrophage polarization through IGF2BP1/IRAK1 axis.
Collapse
Affiliation(s)
- Yue Han
- The Second Department of Gastrointestinal Surgery, Shandong Provincial Third Hospital, Jinan City, China
| | - Zhe Zhou
- The Second Department of Gastrointestinal Surgery, Shandong Provincial Third Hospital, Jinan City, China
| | - Rudong Li
- The Second Department of Gastrointestinal Surgery, Shandong Provincial Third Hospital, Jinan City, China
| | - Hong Wang
- The Second Department of Gastrointestinal Surgery, Shandong Provincial Third Hospital, Jinan City, China
| |
Collapse
|
25
|
Li Q, Xu J, Hua R, Xu H, Wu Y, Cheng X. Nano-strategies for Targeting Tumor-Associated Macrophages in Cancer immunotherapy. J Cancer 2025; 16:2261-2274. [PMID: 40302816 PMCID: PMC12036086 DOI: 10.7150/jca.108194] [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: 12/04/2024] [Accepted: 02/12/2025] [Indexed: 05/02/2025] Open
Abstract
Tumor-associated macrophages (TAMs) are one type of the most abundant immune cells within tumor, resulting in immunosuppresive tumor microenvironment and tumor resistance to immunotherapy. Thus, targeting TAMs is a promising therapeutic strategy for boosting cancer immunotherapy. This study provides an overview of current therapeutic strategies targeting TAMs, which focus on blocking the recruitment of TAMs by tumors, regulating the polarization of TAMs, and directly eliminating TAMs using various nanodrugs, especially with a new categorization based on the specific signaling pathways, such as NF-κB, HIF-1α, ROS, STAT, JNK, PI3K, and Notch involved in their regulatory mechanism. The latest developments of nanodrugs modulating these pathways are discussed in determining the polarization of TAMs and their role in the tumor microenvironment. Despite the challenges in clinical translation and the complexity of nanodrug synthesis, the potential of nanodrugs in enhancing the effectiveness of cancer immunotherapy is worthy of expecting.
Collapse
Affiliation(s)
- Qian Li
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou 215004, P. R. China
| | - Jingwei Xu
- Department of Thoralic Surgery, Suzhou Municipal Hospital Institution, Suzhou 215000, P. R. China
- Department of Thoralic Surgery, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, 215123, P. R. China
| | - Runjia Hua
- Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, P. R. China
| | - Hanye Xu
- Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, P. R. China
| | - Yongyou Wu
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou 215004, P. R. China
| | - Xiaju Cheng
- Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, P. R. China
| |
Collapse
|
26
|
Long J, Liu B, Li J, Ji X, Zhu N, Zhuang X, Wang H, Li L, Chen Y, Li X, Zhao S. Comprehensive analysis of molecular characteristics between primary and breast-derived metastatic ovarian cancer. Transl Cancer Res 2025; 14:1675-1690. [PMID: 40225000 PMCID: PMC11985203 DOI: 10.21037/tcr-24-1441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 01/14/2025] [Indexed: 04/15/2025]
Abstract
Background The molecular basis for the disparities between primary ovarian cancer (POC) and ovarian cancer secondary to breast cancer (OCSTBC) remains poorly understood. This study aimed to explore the different characteristics between them through genomic analysis. Methods We performed differentially expressed genes (DEGs) analysis between POC (n=96) and OCSTBC (n=44) groups with transcriptome data and revealed the enriched biological pathways with Kyoto Encyclopedia of Genes and Genomes (KEGG) and Hallmark gene sets between these two groups. Then, the Microenvironment Cell Populations (MCP)-counter and Cell-type Identification by Estimating Relative Subsets of RNA Transcript (CIBERSORT) algorithms were applied to evaluate the immune infiltration in tumor microenvironment (TME) between them. Finally, we performed the association analysis within single nucleotide polymorphism (SNP) data and obtained some meaningful SNPs and candidate genes for further transcriptomic analysis. Results We identified a total of 13 cancer-related genes including GATA3, FOXA1, CCND1, and TTK between POC (n=96) and OCSTBC (n=44) groups with DEGs analysis. Integrated analysis revealed more significant immune-enriched pathways in the POC than in the OCSTBC group. Most immune cells had higher infiltration abundance in POC, except M2 macrophages, which was higher in OCSTBC. In SNP analysis, four SNP regions (8q12.1, 11q21, 11q24.3, and 17q25.3) were found to be significantly correlated with phenotypes (POC/OCSTBC), and importantly, some new susceptibility genes such as ETS1, CWC15, and XKR4 were revealed to potentially be associated with distinction between POC and OCSTBC. Conclusions Our study provides a systematic molecular characteristic between POC and OCSTBC and suggests a pressing need to develop some specific therapeutic strategies in certain types of ovarian cancer.
Collapse
Affiliation(s)
- Junqi Long
- School of Software Engineering, Faculty of Information Technology, Beijing University of Technology, Beijing, China
| | - Bo Liu
- School of Mathematical and Computational Sciences, Massey University, Auckland, New Zealand
| | - Jinmeng Li
- School of Software Engineering, Faculty of Information Technology, Beijing University of Technology, Beijing, China
| | - Xinchan Ji
- School of Software Engineering, Faculty of Information Technology, Beijing University of Technology, Beijing, China
| | - Nian Zhu
- School of Software Engineering, Faculty of Information Technology, Beijing University of Technology, Beijing, China
| | - Xujie Zhuang
- School of Software Engineering, Faculty of Information Technology, Beijing University of Technology, Beijing, China
| | - Huina Wang
- School of Software Engineering, Faculty of Information Technology, Beijing University of Technology, Beijing, China
| | - Lujia Li
- School of Software Engineering, Faculty of Information Technology, Beijing University of Technology, Beijing, China
| | - Yuhaoran Chen
- School of Software Engineering, Faculty of Information Technology, Beijing University of Technology, Beijing, China
| | - Xiaohui Li
- Department of Joint Surgery, Tianjin Hospital, Tianjin University, Tianjin, China
| | - Shuangtao Zhao
- Department of Thoracic Surgery, Beijing Tuberculosis and Thoracic Tumor Research Institute/Beijing Chest Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
27
|
Jiang B, Yang J, Huang Q, Li W, Peng Q, Gan H, Peng T, Yao L, Qi L. Schisandrin B downregulates exosomal fibronectin 1 expression to inhibit hepatocellular carcinoma growth. Front Pharmacol 2025; 16:1547685. [PMID: 40223922 PMCID: PMC11986357 DOI: 10.3389/fphar.2025.1547685] [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: 12/18/2024] [Accepted: 03/10/2025] [Indexed: 04/15/2025] Open
Abstract
Introduction In recent years, natural compounds have attracted wide attention for the treatment of liver cancer due to their therapeutic potential and reduced toxicity. Among these, Schisandrin B (Sch B), a primary bioactive component derived from Schisandra chinensis, has shown notable antitumor activity; however, its specific mechanism remains unclear. Methods The effect of Sch B on the growth of hepatocellular carcinoma(HCC) cells were assessed using CCK-8 assay, colony formation assay and EdU assay, and apoptosis was detected by flow cytometry. The co-culture system of macrophages and HCC cells was established to detect the effect of Sch B on the cell viability and cell cycle changes of HCC cells in the co-culture system. Then, the migration of HCC cells in the co-culture system was studied using a subtoxic concentration of Sch B. Exosomes of the co-culture system with or without Sch B effect were collected for identification and protein spectrum analysis. The differential protein was analyzed by KEGG enrichment analysis and protein interaction network, which was verified by western blotting. Meanwhile, the expression changes of macrophage polarization markers were detected. Finally, the inhibitory effect of Sch B on HCC and the changes of FN1 were verified by in vivo experiments. Results Sch B inhibited HCC cell growth; moreover, it significantly suppressed HCC cell proliferation in the co-culture system and induced S-phase cell cycle arrest by downregulating CDK4, CDK2, and cyclin A2 while upregulating p27 Kip1. Additionally, Sch B inhibited the migration of HCC cells in the co-culture system.The differentially expressed protein fibronectin 1(FN1) in liver cancer patients was higher than that in healthy people. Moreover, after SchB treatment, the expression of FN1 protein in exosomes decreased and the macrophages exhibited M1 polarization. In vivo experiments also verified that Sch B inhibited HCC growth and downregulated the expression of FN1 protein in tumor tissues. Conclusion Sch B may inhibit the development of HCC by inhibiting the expression of exosomal FN1during interactions between macrophages and HCC cells.
Collapse
Affiliation(s)
- Baoyi Jiang
- Division of Gastroenterology, Institute of Digestive Disease, The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan, Guang Dong, China
| | - Jie Yang
- Division of Gastroenterology, Institute of Digestive Disease, The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan, Guang Dong, China
| | - Qingtian Huang
- Division of Gastroenterology, Institute of Digestive Disease, The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan, Guang Dong, China
- Department of Pathology, The Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People’s Hospital, Qingyuan, Guang Dong, China
| | - Wei Li
- Biological Sample Resource Centre, The Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People’s Hospital, Qingyuan, Guang Dong, China
| | - Qian Peng
- Division of Gastroenterology, Institute of Digestive Disease, The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan, Guang Dong, China
| | - Huoye Gan
- Division of Gastroenterology, Institute of Digestive Disease, The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan, Guang Dong, China
| | - Tieli Peng
- Division of Gastroenterology, Institute of Digestive Disease, The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan, Guang Dong, China
| | - Leyi Yao
- Zhanjiang Institute of Clinical Medicine, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, China
| | - Ling Qi
- Division of Gastroenterology, Institute of Digestive Disease, The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan, Guang Dong, China
- Biological Sample Resource Centre, The Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People’s Hospital, Qingyuan, Guang Dong, China
| |
Collapse
|
28
|
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. [PMID: 40160689 PMCID: PMC11947897 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: 11/25/2024] [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
|
29
|
Li Y, Wang H, Mao D, Che X, Chen Y, Liu Y. Understanding pre-metastatic niche formation: implications for colorectal cancer liver metastasis. J Transl Med 2025; 23:340. [PMID: 40098140 PMCID: PMC11912654 DOI: 10.1186/s12967-025-06328-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 03/01/2025] [Indexed: 03/19/2025] Open
Abstract
The liver is the most commonly metastasized organ in colorectal cancer (CRC), and distant metastasis is the primary cause of mortality from CRC. In recent years, researchers have discovered that tumor cells create a "pre-metastatic niche (PMN)" favorable to metastasis before reaching the metastatic location. This review discusses the many processes and mechanisms that lead to PMN formation in CRC, including gut microbiota, stem cell stimulation, immunocyte interactions, and the induction of extracellular vesicles that carry important information. It examines research methods and diagnostic and therapeutic approaches for treating metastatic CRC with PMN. The crucial significance of PMN formation in metastatic CRC is also highlighted.
Collapse
Affiliation(s)
- Yaqin Li
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, China
- Multi-Component of Traditional Chinese Medicine and MicroecologyResearch Center, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, China
| | - Hong Wang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, China
- Multi-Component of Traditional Chinese Medicine and MicroecologyResearch Center, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, China
| | - Dengxuan Mao
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, China
- Multi-Component of Traditional Chinese Medicine and MicroecologyResearch Center, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, China
| | - Xiaoyu Che
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, China
- Multi-Component of Traditional Chinese Medicine and MicroecologyResearch Center, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, China
| | - Yan Chen
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, China.
- Multi-Component of Traditional Chinese Medicine and MicroecologyResearch Center, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, China.
- Jiangsu Clinical Innovation Center of Digestive Cancer of Traditional Chinese Medicine, Administration of Traditional Chinese Medicine of Jiangsu Province, Nanjing, China.
| | - Yuping Liu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, China.
- Multi-Component of Traditional Chinese Medicine and MicroecologyResearch Center, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, China.
- Jiangsu Clinical Innovation Center of Digestive Cancer of Traditional Chinese Medicine, Administration of Traditional Chinese Medicine of Jiangsu Province, Nanjing, China.
| |
Collapse
|
30
|
Geng JX, Lu YF, Zhou JN, Huang B, Qin Y. Exosome technology: A novel and effective drug delivery system in the field of cancer therapy. World J Gastrointest Oncol 2025; 17:101857. [PMID: 40092946 PMCID: PMC11866225 DOI: 10.4251/wjgo.v17.i3.101857] [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: 09/28/2024] [Revised: 11/23/2024] [Accepted: 12/20/2024] [Indexed: 02/14/2025] Open
Abstract
In this article, we revisit an article, which specifically focuses on the utilization of exosomes derived from human bone marrow mesenchymal stem cells (MSCs) for targeted delivery of gemcitabine in pancreatic cancer treatment. The experimental results demonstrated that the exosome-based drug delivery system derived from MSCs significantly augmented apoptosis in pancreatic cancer cells. The biocompatibility, targeting specificity, and low immunogenicity of exosomes render them as optimal carriers for drug delivery, enabling precise administration of therapeutics to diseased tissues while mitigating adverse effects, thereby achieving targeted treatment of cancer cells and significantly enhancing anti-tumor efficacy. However, the clinical application of exosome drug delivery platforms in oncology still presents challenges, necessitating further optimization to ensure their stability and efficacy. This study focuses on elucidating the advantages of exosomes as a drug delivery platform, exploring the utilization of MSC-derived exosomes in oncology therapy, and discussing their potential and future directions in cancer treatment.
Collapse
Affiliation(s)
- Jia-Xin Geng
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, Zhejiang Province, China
| | - Yao-Fan Lu
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, Zhejiang Province, China
| | - Jing-Nan Zhou
- Zhejiang Cancer Hospital, Hangzhou 310018, Zhejiang Province, China
| | - Biao Huang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, Zhejiang Province, China
| | - Yuan Qin
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, Zhejiang Province, China
| |
Collapse
|
31
|
Qiao S, Li X, Yang S, Hua H, Mao C, Lu W. Investigating the PI3K/AKT/mTOR axis in Buzhong Yiqi Decoction's anti-colorectal cancer activity. Sci Rep 2025; 15:8238. [PMID: 40065054 PMCID: PMC11893811 DOI: 10.1038/s41598-025-89018-9] [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: 11/16/2024] [Accepted: 02/03/2025] [Indexed: 03/14/2025] Open
Abstract
Buzhong Yiqi Decoction (BZYQD) is a traditional Chinese medicine renowned for its anti-colorectal cancer (CRC) properties. However, the bioactive components and mechanisms of BZYQD against CRC remain unknown. In this study, LC-MS was used to analyze the chemical composition of BZYQD. Next, the network pharmacology and molecular docking was used to investigate the core components and targets of BZYQD against CRC. Finally, we experimentally validated the potential mechanism of BZYQD against CRC through in vitro studies. Our results identified 26 chemical components in the BZYQD; 75 "hithubs" targets were screened by network pharmacology, and mainly involving pathways such as including pathways in cancer, P13K-Akt signaling pathway, proteoglycans in cancer, kaposi sarcoma-associated herpesvirus, and lipid and atherosclerosis signaling pathways. Based on the number of "hithubs" targets in the key pathways, the two most critical targets including AKT1 and PIK3CA were selected. The component-target network results indicated that astragaloside IV, gancaonin A, quercetin, poricoic acid A, and licoisoflavanone are key anti-CRC components in BZYQD. Molecular docking showed a strong binding affinity between these components and targets. The phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) signaling pathway emerged as the primary target of BZYQD. Further in vitro studies confirmed that BZYQD's anti-CRC activity is mediated through the PI3K/AKT/mTOR axis and influences macrophage polarization. BZYQD exerts its therapeutic effects on CRC through multiple components, targets, and pathways. Our study elucidates the effective components and molecular mechanisms of BZYQD in CRC treatment and provides preliminary validation through molecular docking and experimental studies.
Collapse
Affiliation(s)
- Song Qiao
- Department of Oncology and Hematology, Xijing 986 Hospital, No. 269 Youyi East Road, Beilin District, Xi'an, 710054, Shaanxi, China
| | - Xiaolong Li
- Radiotherapy 1 Ward, Shaanxi Provincial Cancer Hospital, No. 309, Yanta West Road, Yanta District, Xi'an, Shaanxi, China
| | - Shangzhen Yang
- Department of Oncology and Hematology, Xijing 986 Hospital, No. 269 Youyi East Road, Beilin District, Xi'an, 710054, Shaanxi, China
| | - Hua Hua
- Department of Oncology and Hematology, Xijing 986 Hospital, No. 269 Youyi East Road, Beilin District, Xi'an, 710054, Shaanxi, China
| | - Chengtao Mao
- Department of Oncology and Hematology, Xijing 986 Hospital, No. 269 Youyi East Road, Beilin District, Xi'an, 710054, Shaanxi, China
| | - Wanling Lu
- Department of Oncology and Hematology, Xijing 986 Hospital, No. 269 Youyi East Road, Beilin District, Xi'an, 710054, Shaanxi, China.
| |
Collapse
|
32
|
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
|
33
|
Tian J, Zhang L, La X, Fan X, Li Z. MiR-769-5p of macrophage exosomes induced by GRP78 promotes stemness and chemoresistance in colorectal cancer. Cell Death Dis 2025; 16:156. [PMID: 40044682 PMCID: PMC11882909 DOI: 10.1038/s41419-025-07466-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 02/03/2025] [Accepted: 02/19/2025] [Indexed: 03/09/2025]
Abstract
The tumor microenvironment (TME) plays an important role in tumorigenesis and development. Tumor-associated macrophages (TAMs) are essential members of the TME, the exosomes and miRNAs they secrete are crucial in tumor regulation. Our previous study showed that GRP78-induced macrophages infinitely tend to be M2-type TAMs. In this study, the exosomes of M0 and GRP78-induced macrophage were collected and co-incubated with colorectal cancer (CRC) cells. The results implied that macrophage exosomes induced by GRP78 (GRP78-exos) significantly promoted stemness and chemoresistance in CRC in vitro and in vivo. Further, the top 5 miRNAs upregulated in GRP78-exos were obtained from miRNA sequencing data. The qRT-PCR validation revealed that miR-769-5p was the most observably upregulated and could be directly transferred into CRC cells via GRP78-exos. Mechanistically, the study indicated that miR-769-5p targeted MAPK1 to regulate the cell cycle-related proteins RB1, cyclin D1, and cyclin E1. This contributes to CRC cells entering a quiescent state, which leads to the development of chemoresistance. Moreover, miR-769-5p is also expressed higher in the tissues of 5-FU-resistant CRC patients. In summary, the findings indicate a novel function of miR-769-5p as a potential marker for the diagnosis and treatment of chemotherapy resistance in CRC.
Collapse
Affiliation(s)
- Jinmiao Tian
- Key Laboratory of Chemical Biology and Molecular Engineering of the National Ministry of Education, Institute of Biotechnology, Shanxi University, Taiyuan, 030006, China
| | - Lichao Zhang
- Institutes of Biomedical Sciences, Shanxi University, Taiyuan, 030006, China.
| | - Xiaoqin La
- Institutes of Biomedical Sciences, Shanxi University, Taiyuan, 030006, China
| | - Xiaxia Fan
- Key Laboratory of Chemical Biology and Molecular Engineering of the National Ministry of Education, Institute of Biotechnology, Shanxi University, Taiyuan, 030006, China
| | - Zhuoyu Li
- Key Laboratory of Chemical Biology and Molecular Engineering of the National Ministry of Education, Institute of Biotechnology, Shanxi University, Taiyuan, 030006, China.
| |
Collapse
|
34
|
Liu R, Tang L, Liu Y, Hu H, Liu J. Causal relationship between immune cell signatures and colorectal cancer: a bi-directional, two-sample mendelian randomization study. BMC Cancer 2025; 25:387. [PMID: 40033246 PMCID: PMC11877943 DOI: 10.1186/s12885-025-13576-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 01/21/2025] [Indexed: 03/05/2025] Open
Abstract
BACKGROUND Prior studies have demonstrated the association between immune cells and colorectal cancer (CRC). However, the causal link to specific immunophenotypes is limited. This study intends to elucidate the causal relationship of immune cell signatures on CRC. METHODS We performed a bi-directional and two-sample mendelian randomization (MR) study, utilizing GWAS summary data of 731 immune cell traits (n = 3,757) and CRC statistics (n = 470,002). The primary MR methodology was inverse-variance weighted (IVW) method. Furthermore, heterogeneity was evaluated by Cochran's Q test. MR-PRESSO and MR-Egger were employed to assess horizontal and vertical pleiotropy respectively. Sensitivity analysis and FDR correction were conducted in our results. These results were validated in both the UK Biobank and FinnGen cohorts. We also extracted transcriptomic data of CRC and adjacent non-tumor tissues from TCGA, and used CIBERSORT to compare the infiltration patterns of 22 immune cell panels between normal tissues and the tumor microenvironment (TME). RESULTS Our study indicated nine immune cell signatures had significant causality with the risk of CRC after sensitivity analysis and FDR correction. The positive results covered four panels: B cell, CD8 + T cell, Treg, and monocyte. IgD- CD38br and IgD + CD38br B cell, CD8dim and CD28 + CD45RA- CD8dim T cell, and CD14 on CD14 + CD16- monocyte were the protective factors of CRC. However, CD39 + resting Treg, CX3CR1 on CD14- CD16 + monocyte, FSC-A on HLA DR + T cell, and BAFF-R on B cell increased the risk of CRC. The results were validated in the UK Biobank data and FinnGen cohorts. The data from the TCGA database also confirmed the infiltration of B cell, CD8 + T cell, Treg, and monocyte panels in the TME. CONCLUSION This study highlights the causal link between specific immune cell phenotypes and CRC, providing valuable insights into the immune microenvironment's role in CRC. The validation of our findings using large-scale datasets (UK Biobank, FinnGen) and TCGA underscores the robustness of our results, offering new potential therapeutic targets for CRC treatment.
Collapse
Affiliation(s)
- Ruizhi Liu
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Liansha Tang
- Department of Biotherapy, Cancer Center, West China Hospital of Sichuan University, 37 Guoxue Xiang Street, Chengdu, Sichuan Province, 610041, China
| | - Yunjia Liu
- Mental Health Center and Psychiatric Laboratory, The State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Handan Hu
- Queen Mary College, Nanchang University, Nanchang, Jiangxi, China
| | - Jiyan Liu
- Department of Biotherapy, Cancer Center, West China Hospital of Sichuan University, 37 Guoxue Xiang Street, Chengdu, Sichuan Province, 610041, China.
| |
Collapse
|
35
|
Liu X, To KK, Zeng Q, Fu L. Effect of Extracellular Vesicles Derived From Tumor Cells on Immune Evasion. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2417357. [PMID: 39899680 PMCID: PMC11948033 DOI: 10.1002/advs.202417357] [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: 12/21/2024] [Indexed: 02/05/2025]
Abstract
The crosstalk between immunity and cancer in the regulation of tumor growth is considered a hallmark of cancer. Antitumor immunity refers to the innate and adaptive immune responses that regulate cancer development and proliferation. Tumor immune evasion represents a major hindrance to effective anticancer treatment. Extracellular vesicles (EVs) are nano-sized and lipid-bilayer-enclosed particles that are secreted to the extracellular space by all cell types. They are critically involved in numerous biological functions including intercellular communication. Tumor-derived extracellular vesicles (TEVs) can transport a variety of cargo to modulate immune cells in the tumor microenvironment (TME). This review provides the latest update about how tumor cells evade immune surveillance by exploiting TEVs. First, the biogenesis of EVs and the cargo-sorting machinery are discussed. Second, how tumor cells modulate immune cell differentiation, activation, and function via TEVs to evade immune surveillance is illustrated. Last but not least, the novel antitumor strategies that can reverse immune escape are summarized.
Collapse
Affiliation(s)
- Xuanfan Liu
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer MedicineGuangdong Esophageal Cancer InstituteSun Yat‐sen University Cancer CenterGuangzhou510060P. R. China
- Department of UrologyThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhou510080P. R. China
| | - Kenneth K.W. To
- School of PharmacyThe Chinese University of Hong KongHong Kong999077P. R. China
| | - Qinsong Zeng
- Department of UrologyThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhou510080P. R. China
- Guangxi Hospital Division of The First Affiliated HospitalSun Yat‐sen UniversityNanning530025P. R. China
| | - Liwu Fu
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer MedicineGuangdong Esophageal Cancer InstituteSun Yat‐sen University Cancer CenterGuangzhou510060P. R. China
| |
Collapse
|
36
|
Jiang M, Zhang K, Meng J, Xu L, Liu Y, Wei R. Engineered exosomes in service of tumor immunotherapy: From optimizing tumor-derived exosomes to delivering CRISPR/Cas9 system. Int J Cancer 2025; 156:898-913. [PMID: 39474936 DOI: 10.1002/ijc.35241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 10/09/2024] [Accepted: 10/17/2024] [Indexed: 01/07/2025]
Abstract
Exosomes can be modified and designed for various therapeutic goals because of their unique physical and chemical characteristics. Researchers have identified tumor-derived exosomes (TEXs) as significant players in cancer by influencing tumor growth, immune response evasion, angiogeneis, and drug resistance. TEXs promote the production of specific proteins important for cancer progression. Due to their easy accessibility, TEXs are being modified through genetic, drug delivery, membrane, immune system, and chemical alterations to be repurposed as vehicles for delivering drugs to improve cancer treatment outcomes. In the complex in vivo environment, the clustered regularly interspaced short palindromic repeats and CRISPR-associated protein 9 (CRISPR/Cas9) system encounters challenges from degradation, neutralization, and immune responses, emphasizing the need for strategic distribution strategies for effective genome editing. Engineered exosomes present a promising avenue for delivering CRISPR/Cas9 in vivo. In this review, we will explore different techniques for enhancing TEXs using various engineering strategies. Additionally, we will discuss how these exosomes can be incorporated into advanced genetic engineering systems like CRISPR/Cas9 for possible therapeutic uses.
Collapse
Affiliation(s)
- Mingyang Jiang
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Ke Zhang
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jinfeng Meng
- The First Clinical Medical College, Guangxi Medical University, Nanning
| | - Linhua Xu
- The First Clinical Medical College, Guangxi Medical University, Nanning
| | - Ying Liu
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Ruqiong Wei
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
37
|
Shao S, Tang L, Rong W, Han Y, Liu X, Zhu H, Song Q, Ji Q. Bushen Jiedu formula alleviates colorectal cancer progression through reducing lncRPPH1 in tumor-derived extracellular vesicles. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 138:156390. [PMID: 39884077 DOI: 10.1016/j.phymed.2025.156390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 12/26/2024] [Accepted: 01/12/2025] [Indexed: 02/01/2025]
Abstract
BACKGROUND The Bushen Jiedu Formula (BSJDF) is a traditional and effective chemical prescription of traditional Chinese medicine (TCM) administered due to its anti-cancer properties, particularly in colorectal cancer (CRC). PURPOSE This study proposes to explore the therapeutic benefits of BSJDF against metastasis in CRC and unravel its regulatory mechanisms related to the tumor microenvironment. STUDY DESIGN/METHODS The combination of mass spectrometry and network pharmacology was used to analyze the involvement of BSJDF in anti-tumor progression. In vitro and in vivo experiments were conducted to measure the regulatory effect of BSJDF on tumor-derived extracellular vesicles (EVs), which induce the M2 polarization of macrophages and CRC metastasis. Flow cytometry, immunofluorescence, and RT-qPCR assays were employed to elucidate the mechanisms by which tumor-derived EVs induce macrophage M2-type polarization. RESULTS Network pharmacology illuminated that immune and inflammatory response pathways were involved in the beneficial effects of BSJDF on CRC. In vivo experiments indicated that BSJDF suppressed the metastasis of CRC to the liver by modulating macrophage immune infiltration. Mechanically, BSJDF inhibited CRC metastasis via modulating tumor-derived EVs that facilitate the polarization of M2 macrophages. Moreover, BSJDF suppressed the metastasis of CRC and the polarization of M2 macrophages by reducing lncRPPH1 in tumor-derived EVs. CONCLUSIONS BSJDF blocked the M2-type polarization of macrophages and prevented CRC metastasis by decreasing the expression levels of lncRPPH1 in tumor-derived EVs.
Collapse
Affiliation(s)
- Shiyun Shao
- Department of Medical Oncology & Cancer Institute of Integrative Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Lei Tang
- Department of Medical Oncology & Cancer Institute of Integrative Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Wenqing Rong
- Department of Medical Oncology & Cancer Institute of Integrative Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Department of Medical Oncology, Seventh People's Hospital of Shanghai University of TCM, Shanghai 200137, China.
| | - Yicun Han
- Department of Medical Oncology & Cancer Institute of Integrative Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Xiaodie Liu
- Department of Medical Oncology & Cancer Institute of Integrative Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Huirong Zhu
- Department of Medical Oncology & Cancer Institute of Integrative Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Qing Song
- Department of Medical Oncology, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou 215007, Jiangsu, China.
| | - Qing Ji
- Department of Medical Oncology & Cancer Institute of Integrative Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
38
|
Li Z, Deng L, Cheng M, Ye X, Yang N, Fan Z, Sun L. Emerging role of bile acids in colorectal liver metastasis: From molecular mechanism to clinical significance (Review). Int J Oncol 2025; 66:24. [PMID: 39981904 PMCID: PMC11844338 DOI: 10.3892/ijo.2025.5730] [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: 10/25/2024] [Accepted: 01/20/2025] [Indexed: 02/22/2025] Open
Abstract
Liver metastasis is the leading cause of colorectal cancer (CRC)‑related mortality. Microbiota dysbiosis serves a role in the pathogenesis of colorectal liver metastases. Bile acids (BAs), cholesterol metabolites synthesized by intestinal bacteria, contribute to the metastatic cascade of CRC, encompassing colorectal invasion, migration, angiogenesis, anoikis resistance and the establishment of a hepatic pre‑metastatic niche. BAs impact inflammation and modulate the immune landscape within the tumor microenvironment by activating signaling pathways, which are used by tumor cells to facilitate metastasis. Given the widespread distribution of BA‑activated receptors in both tumor and immune cells, strategies aimed at restoring BA homeostasis and blocking metastasis‑associated signaling are of importance in cancer therapy. The present study summarizes the specific role of BAs in each step of colorectal liver metastasis, elucidating the association between BA and CRC progression to highlight the potential of BAs as predictive biomarkers for colorectal liver metastasis and their therapeutic potential in developing novel treatment strategies.
Collapse
Affiliation(s)
- Zhaoyu Li
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, P.R. China
| | - Lingjun Deng
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China, P.R. China
| | - Mengting Cheng
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China, P.R. China
| | - Xiandong Ye
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China, P.R. China
| | - Nanyan Yang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China, P.R. China
| | - Zaiwen Fan
- Department of Oncology, Air Force Medical Center of People's Liberation Army, Air Force Medical University, Beijing 100010, P.R. China
| | - Li Sun
- Department of Oncology, Air Force Medical Center of People's Liberation Army, Air Force Medical University, Beijing 100010, P.R. China
| |
Collapse
|
39
|
Xu W, Xu J, Liu J, Wang N, Zhou L, Guo J. Liver Metastasis in Cancer: Molecular Mechanisms and Management. MedComm (Beijing) 2025; 6:e70119. [PMID: 40027151 PMCID: PMC11868442 DOI: 10.1002/mco2.70119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 01/15/2025] [Accepted: 01/20/2025] [Indexed: 03/05/2025] Open
Abstract
Liver metastasis is a leading cause of mortality from malignant tumors and significantly impairs the efficacy of therapeutic interventions. In recent years, both preclinical and clinical research have made significant progress in understanding the molecular mechanisms and therapeutic strategies of liver metastasis. Metastatic tumor cells from different primary sites undergo highly similar biological processes, ultimately achieving ectopic colonization and growth in the liver. In this review, we begin by introducing the inherent metastatic-friendly features of the liver. We then explore the panorama of liver metastasis and conclude the three continuous, yet distinct phases based on the liver's response to metastasis. This includes metastatic sensing stage, metastatic stress stage, and metastasis support stage. We discuss the intricate interactions between metastatic tumor cells and various resident and recruited cells. In addition, we emphasize the critical role of spatial remodeling of immune cells in liver metastasis. Finally, we review the recent advancements and the challenges faced in the clinical management of liver metastasis. Future precise antimetastatic treatments should fully consider individual heterogeneity and implement different targeted interventions based on stages of liver metastasis.
Collapse
Affiliation(s)
- Wenchao Xu
- Department of General SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Key Laboratory of Research in Pancreatic TumorChinese Academy of Medical SciencesBeijingChina
- National Infrastructures for Translational MedicinePeking Union Medical College HospitalBeijingChina
- State Key Laboratory of ComplexSevere, and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Jia Xu
- State Key Laboratory of Fine ChemicalsDepartment of Pharmaceutical SciencesSchool of Chemical EngineeringDalian University of TechnologyDalianChina
| | - Jianzhou Liu
- Department of General SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Key Laboratory of Research in Pancreatic TumorChinese Academy of Medical SciencesBeijingChina
- National Infrastructures for Translational MedicinePeking Union Medical College HospitalBeijingChina
- State Key Laboratory of ComplexSevere, and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Nanzhou Wang
- Department of Colorectal SurgeryState Key Laboratory of Oncology in South ChinaSun Yat‐sen University Cancer CenterGuangdong Provincial Clinical Research Center for CancerGuangzhouChina
| | - Li Zhou
- Department of General SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Key Laboratory of Research in Pancreatic TumorChinese Academy of Medical SciencesBeijingChina
- National Infrastructures for Translational MedicinePeking Union Medical College HospitalBeijingChina
- State Key Laboratory of ComplexSevere, and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Junchao Guo
- Department of General SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Key Laboratory of Research in Pancreatic TumorChinese Academy of Medical SciencesBeijingChina
- National Infrastructures for Translational MedicinePeking Union Medical College HospitalBeijingChina
- State Key Laboratory of ComplexSevere, and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
40
|
Li C, Liu C, Ma H, Zhang Z, Zhang J. Lymphocytes-Associated Extracellular Vesicles Activate Natural Killer Cells in HNSCC. Cancer Sci 2025; 116:633-642. [PMID: 39749376 PMCID: PMC11875761 DOI: 10.1111/cas.16440] [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: 08/09/2024] [Revised: 12/05/2024] [Accepted: 12/13/2024] [Indexed: 01/04/2025] Open
Abstract
Small extracellular vesicles (sEVs) facilitate intercellular communication and play a pivotal role in tumor progression. Accumulated evidence has indicated the diversity of sEVs but with limited results revealing the landscape of heterogeneity of sEVs. The heterogeneity of cargo RNA in sEVs presents the different cell origins and indicates different functions. Here, we analyzed the heterogeneity of sEVs at droplet levels from single-cell RNA sequencing results of head and neck squamous cell carcinoma (HNSCC) with the previously reported algorithm SEVtras. With the sEVs secretion activity calculated by SEVtras, we also found that the T cells held the major role of sEVs secretion. In addition, we found these sEVs secreted by T cells increased the cytotoxic ability of natural killer cells (NK cells), which illustrated an indirect manner for the anti-tumor function of T cells. These results revealed the heterogeneity of cargo RNA of sEVs in HNSCC and underlined a sEVs-dependent manner in which T cells act on NK cells and anti-tumor immunity.
Collapse
Affiliation(s)
- Chuwen Li
- Department of Oral and Maxillofacial‐Head and Neck Oncology, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiPeople's Republic of China
- College of StomatologyShanghai Jiao Tong UniversityShanghaiPeople's Republic of China
- National Center for StomatologyShanghaiPeople's Republic of China
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of StomatologyShanghai Center of Head and Neck Oncology Clinical and Translational ScienceShanghaiPeople's Republic of China
| | - Chun Liu
- Department of Oral and Maxillofacial‐Head and Neck Oncology, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiPeople's Republic of China
- College of StomatologyShanghai Jiao Tong UniversityShanghaiPeople's Republic of China
- National Center for StomatologyShanghaiPeople's Republic of China
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of StomatologyShanghai Center of Head and Neck Oncology Clinical and Translational ScienceShanghaiPeople's Republic of China
| | - Hailong Ma
- Department of Oral and Maxillofacial‐Head and Neck Oncology, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiPeople's Republic of China
- College of StomatologyShanghai Jiao Tong UniversityShanghaiPeople's Republic of China
- National Center for StomatologyShanghaiPeople's Republic of China
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of StomatologyShanghai Center of Head and Neck Oncology Clinical and Translational ScienceShanghaiPeople's Republic of China
| | - Zhiyuan Zhang
- Department of Oral and Maxillofacial‐Head and Neck Oncology, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiPeople's Republic of China
- College of StomatologyShanghai Jiao Tong UniversityShanghaiPeople's Republic of China
- National Center for StomatologyShanghaiPeople's Republic of China
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of StomatologyShanghai Center of Head and Neck Oncology Clinical and Translational ScienceShanghaiPeople's Republic of China
- Research Unit of Oral and Maxillofacial Regenerative MedicineChinese Academy of Medical SciencesShanghaiPeople's Republic of China
| | - Jianjun Zhang
- Department of Oral and Maxillofacial‐Head and Neck Oncology, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiPeople's Republic of China
- College of StomatologyShanghai Jiao Tong UniversityShanghaiPeople's Republic of China
- National Center for StomatologyShanghaiPeople's Republic of China
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of StomatologyShanghai Center of Head and Neck Oncology Clinical and Translational ScienceShanghaiPeople's Republic of China
| |
Collapse
|
41
|
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
|
42
|
Liu W, Qiao D, Chen J, Gao Y, Okuda K, Shimada Y, Yao L. Aspirin impedes non-small cell lung cancer development via fine-tuning the CD36 localization regulated by GPIHBP1. Transl Lung Cancer Res 2025; 14:491-512. [PMID: 40114952 PMCID: PMC11921230 DOI: 10.21037/tlcr-2024-1174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 02/19/2025] [Indexed: 03/22/2025]
Abstract
Background Lung cancer, a commonly diagnosed malignancy, is the leading cause of cancer-related death worldwide. Aspirin suppresses the progression and metastasis of various cancers. However, the effect of aspirin on non-small cell lung cancer (NSCLC) has not been fully understood. It has been established that glycosylphosphatidylinositol HDL-binding protein 1 (GPIHBP1) and CD36 play a vital role in lipid metabolism and transport. This study aimed to clarify the mechanism by which aspirin inhibits NSCLC cell proliferation and metastasis via GPIHBP1. Methods The blood and tissues of 10 patients with NSCLC treated with aspirin and 10 patients without aspirin were collected and analyzed via RNA sequencing. GPIHBP1 expression was determined by immunohistochemistry (IHC), Western blotting, and quantitative real time polymerase chain reaction (qRT-PCR). A series of functional experiments were performed to evaluate the effects of aspirin on NSCLC progression in a GPIHBP1-dependent manner. The potential mechanism of GPIHBP1 was explored via coimmunoprecipitation and immunofluorescence staining. The effect of GPIHBP1 on tumor growth and metastasis was verified by constructing subcutaneous xenograft tumor model in nude mice. Results GPIHBP1 was downregulated and was increased by treatment with aspirin in lung cancer tissues. Furthermore, GPIHBP1 overexpression inhibited the migration, cell proliferation, and epithelial-mesenchymal transition process in NSCLC cells while promoting their apoptosis, while in cells with GPIHBP1 knockdown, the opposite was observed. Mechanistically, GPIHBP1 directly interacted with CD36 while GPIHBP1 knockdown disrupted CD36 localization, thus promoting tumor progression and metastasis in NSCLC cells. In addition, through in vivo xenograft experiments, we found that GPIHBP1 overexpression inhibited tumor growth and metastasis. Conclusions Our findings provide new insights into the mechanism by which aspirin suppresses lung cancer development in a GPIHBP1-dependent manner and may provide a promising target in NSCLC treatment.
Collapse
Affiliation(s)
- Wei Liu
- Department of Critical Care Medicine, Tangdu Hospital of Air Force Military Medical University, Xi'an, China
| | - Dujuan Qiao
- Department of Anesthesiology & Perioperative Medicine (D), Xi'an People's Hospital (Xi'an Fourth Hospital), Xi'an, China
| | - Jia Chen
- The College of Life Sciences, Northwest University, Xi'an, China
| | - Ya Gao
- Department of Critical Care Medicine, Tangdu Hospital of Air Force Military Medical University, Xi'an, China
| | - Katsuhiro Okuda
- Department of Thoracic and Pediatric Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Yoshihisa Shimada
- Department of Thoracic Surgery, Tokyo Medical University, Tokyo, Japan
| | - Linong Yao
- Department of Critical Care Medicine, Tangdu Hospital of Air Force Military Medical University, Xi'an, China
| |
Collapse
|
43
|
Wang W, Yin J. Exosomal miR-203 from bone marrow stem cells targets the SOCS3/NF-κB pathway to regulate neuroinflammation in temporal lobe epilepsy. World J Stem Cells 2025; 17:101395. [DOI: 10.4252/wjsc.v17.i2.101395] [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: 11/28/2024] [Revised: 12/27/2024] [Accepted: 02/11/2025] [Indexed: 02/24/2025] Open
Abstract
BACKGROUND Epilepsy is a prevalent chronic neurological disorder affecting 50 million individuals globally, with temporal lobe epilepsy (TLE) being the most common form. Despite advances in antiepileptic drug development, over 30% of patients suffer from drug-resistant epilepsy, which can lead to severe cognitive impairments and adverse psychosocial outcomes.
AIM To explore the role of bone marrow mesenchymal stem cell (BMSC)-derived exosomal miR-203 in the regulation of neuroinflammation in a mouse model of epilepsy, providing a theoretical basis for the development of targeted microRNA delivery therapies for drug-resistant epilepsy.
METHODS Adult male C57BL/6 mice were divided into a control group and a TLE model of 30 mice each, and the TLE model group was established by injecting kainic acid. BMSCs were isolated from the mice, and exosomes were purified using ultracentrifugation. Exosomal miR-203 was identified and characterized using high-throughput sequencing and quantitative reverse-transcription polymerase chain reaction. The uptake of exosomes by hippocampal neurons and the subsequent effects on neuroinflammatory markers were assessed using in vitro cell culture models.
RESULTS Exosomal miR-203 exhibited a significant upregulation in BMSCs derived from epileptic mice. In vitro investigations demonstrated the efficient internalization of these exosomes by hippocampal neurons, resulting in downregulation of suppressor of cytokine signaling 3 expression and activation of the nuclear factor kappaB pathway, ultimately leading to enhanced secretion of pro-inflammatory cytokines.
CONCLUSION Our study identifies exosomal miR-203 as a key regulator of neuroinflammation in a mouse model of epilepsy. The findings suggest that targeting miR-203 may offer a novel therapeutic strategy for epilepsy by modulating the suppression of cytokine signaling 3/nuclear factor kappaB pathway, thus providing a potential avenue for the development of cell-free therapeutics.
Collapse
Affiliation(s)
- Wei Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Dalian Medical University, Dalian 116023, Liaoning Province, China
| | - Jian Yin
- Department of Neurosurgery, The Second Affiliated Hospital of Dalian Medical University, Dalian 116023, Liaoning Province, China
| |
Collapse
|
44
|
Sun J, Luo J, Liu J, Wu H, Li Y, Xu Y, Liu L, Liu X, Zhang Q. Cancer-secreted exosomal miR-1825 induces angiogenesis to promote colorectal cancer metastasis. Cancer Cell Int 2025; 25:63. [PMID: 39987450 PMCID: PMC11847347 DOI: 10.1186/s12935-025-03674-5] [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: 10/12/2024] [Accepted: 02/04/2025] [Indexed: 02/24/2025] Open
Abstract
BACKGROUND Angiogenesis is one of the important factors related to tumorigenesis, invasion, and metastasis. Cancer-secreted exosomes are essential mediators of intercellular cross-talk and participate in angiogenesis and metastasis. Unveiling the mechanism of angiogenesis is an important way to develop anti-angiogenesis therapeutic strategies to against cancer progression. METHODS miR-1825 expression and relationship with microvascular density were validated in colorectal cancer (CRC) by in situ hybridization (ISH) staining and immunohistochemistry (IHC). Sequential differential centrifugation, transmission electron microscopy, and western blotting analysis were used to extract and characterize exosomes. The effort of exosomal miR-1825 on endothelial cells was examined by transwell assay, wound healing assay, tube formation assay, and aortic ring assay. The relationship of miR-1825, ING1 and the downstream pathway were analyzed by western blot, RT-PCR, Immunofluorescence, and dual-luciferase reporter system analysis. RESULTS Exosomal miR-1825 is associated with angiogenesis in CRC and is enriched in exosomes extracted from the serum of CRC patients. The CRC-secreted exosomal miR-1825 can be transferred into vascular endothelial cells, promoting endothelial cell migration and tube formation in vitro, and facilitating angiogenesis and tumor metastasis in vivo. Mechanistically, exosomal miR-1825 regulates angiogenesis and tumor metastasis by suppressing inhibitor of growth family member 1 (ING1) and activating the TGF-β/Smad2/Smad3 signaling pathway in the recipient HUVECs. CONCLUSIONS Our study demonstrated the CRC-secreted exosomal miR-1825 could be transferred to vascular endothelial cells, subsequently leads to the inhibition of ING1 and the activation of the TGF-β/Smad2/Smad3 signaling pathway, thereby promoting angiogenesis and liver metastasis in CRC. Exosomal miR-1825 is thus a potential diagnostic and therapeutic target for CRC patients.
Collapse
Affiliation(s)
- Jingbo Sun
- Department of Pathology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, Guangdong, China
- Department of General Surgery, The Third Affiliated Hospital of Southern Medical University, 183 West Zhongshan Avenue, Guangzhou, 510630, Guangdong, People's Republic of China
| | - Junjie Luo
- Department of General Surgery, The Third Affiliated Hospital of Southern Medical University, 183 West Zhongshan Avenue, Guangzhou, 510630, Guangdong, People's Republic of China
| | - Jialong Liu
- Department of General Surgery, The Third Affiliated Hospital of Southern Medical University, 183 West Zhongshan Avenue, Guangzhou, 510630, Guangdong, People's Republic of China
| | - Hongmei Wu
- Department of Pathology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, Guangdong, China
| | - Yanyan Li
- Department of Pathology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, Guangdong, China
| | - Yangwei Xu
- Department of Pathology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, Guangdong, China
| | - Lixin Liu
- Department of General Surgery, The Third Affiliated Hospital of Southern Medical University, 183 West Zhongshan Avenue, Guangzhou, 510630, Guangdong, People's Republic of China.
| | - Xiaolong Liu
- Department of General Surgery, The Third Affiliated Hospital of Southern Medical University, 183 West Zhongshan Avenue, Guangzhou, 510630, Guangdong, People's Republic of China.
| | - Qingling Zhang
- Department of Pathology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, Guangdong, China.
| |
Collapse
|
45
|
Wang J, Zhu Y, He Y, Shao W. TAM-derived exosomal miR-589-3p accelerates ovarian cancer progression through BCL2L13. J Ovarian Res 2025; 18:36. [PMID: 39985077 PMCID: PMC11846191 DOI: 10.1186/s13048-025-01618-1] [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: 07/09/2024] [Accepted: 02/05/2025] [Indexed: 02/24/2025] Open
Abstract
BACKGROUND Tumor-associated macrophages (TAM) are critical elements of intercellular communication in tumor microenvironment (TME), and exosomes are key mediators between tumor cells and the TME. According to previous reports, miRNAs exert a pivotal role in ovarian cancer (OC) development. The purpose of this work was to explore the function of TAM-derived exosomal miR-589-3p in OC development and elucidate the underlying molecular mechanisms. METHODS First, peripheral blood mononuclear cells (PBMC) were treated with IL-4 and IL-13 to polarize them into M2-type macrophages. Exosomes were separated from M2-type macrophages, and the physical properties of exosomes were evaluated using transmission electron microscopy (TEM) and nanoparticle tracking analysis (NTA). Next, quantitative reverse-transcriptase polymerase chain reaction (qRT-PCR) was applied to examine the expression of relevant genes. Subsequently, Targetscan and miRDB were utilized to predict miR-589-3p target genes, and then the interaction between miR-589-3p and BCL2L13 was verified by dual luciferase assay and RNA Binding Protein Immunoprecipitation (RIP) assay. Finally, Cell Counting Kit-8 (CCK-8) and flow cytometry experiments were employed to explore the changes in the proliferative and apoptotic abilities of OC cells. RESULTS In this research, we demonstrated that TAM-derived exosomes facilitated OC cell proliferation and suppressed OC cell apoptosis. Then, qRT-PCR results indicated that miR-589-3p were markedly elevated after co-culture of TAM-derived exosomes with OC cells. In addition, we discovered that miR-589-3p was bound to BCL-2-like protein 13 (BCL2L13), which was confirmed through luciferase assay and RIP assay. Furthermore, functional analysis displayed that TAM-derived exosomes treated with miR-589-3p inhibitor attenuated the promotion of OC cell progression by exosomes. CONCLUSION TAM-derived exosomal miR-589-3p enhanced OC progression through BCL2L13, which offers a novel for OC therapy.
Collapse
Affiliation(s)
- Jianqing Wang
- Department of Gynecology and Obstetrics, Yancheng First People's Hospital, Yancheng Clinical College of Xuzhou Medical University, Yancheng, Jiangsu, 224002, China
| | - Yan Zhu
- Department of Gynecology and Obstetrics, Yancheng First People's Hospital, Yancheng Clinical College of Xuzhou Medical University, Yancheng, Jiangsu, 224002, China
| | - Yang He
- Department of Gynecology and Obstetrics, Yancheng First People's Hospital, Yancheng Clinical College of Xuzhou Medical University, Yancheng, Jiangsu, 224002, China
| | - Weiwei Shao
- Department of Pathology, Yancheng First People's Hospital, Yancheng Clinical College of Xuzhou Medical University, Yancheng, Jiangsu, 224002, China.
- Department of Pathology, Yancheng Clinical College of Xuzhou Medical University, Yancheng First People's Hospital, No. 166, Yulong West Road, Yancheng, Jiangsu, 224002, China.
| |
Collapse
|
46
|
Liu X, Liu J, Wang X, Zou Y, Tao X, Li J, Ye M, Xu W, Deng Y, Liu L, Sun J, Zhang Q. Cancer-secreted exosomal miR-1246 promotes colorectal cancer liver metastasis by activating hepatic stellate cells. Mol Med 2025; 31:68. [PMID: 39979806 PMCID: PMC11841005 DOI: 10.1186/s10020-025-01112-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 01/31/2025] [Indexed: 02/22/2025] Open
Abstract
Colorectal cancer liver metastases (CRLM) are the primary cause of mortality in colorectal cancer (CRC) patients, highlighting the importance of understanding the underlying mechanisms. The tumor microenvironment (TME) and its interaction with tumor cells play a crucial role in CRLM progression. Notably, the stability and peak levels of tumor-derived exosomal miRNAs facilitate intercellular communication in the TME. Hepatic stellate cells (HSCs), key liver mesenchymal cells, constitute about 33% of the liver's nonsolid cell population and exhibit plasticity. However, the specific role of tumor-derived exosomal miRNAs in the crosstalk between HSCs and tumor cells during the CRLM process remains unclear. We studied CRC-secreted exosomal miR-1246 and its impact on HSCs, as well as its effects on CRC cell proliferation and metastasis. Our findings demonstrate that CRC-secreted exosomal miR-1246 can be internalized by HSCs, leading to their activation and facilitating the metastatic potential of CRC cells. Mechanistically, exosomal miR-1246 targets INSIG1, resulting in SREBP2 nucleation and cholesterol metabolism alterations. This accumulation of free cholesterol (FC) regulates the TLR4/NF-κB/TGF-β pathway, promoting HSC activation. Activated HSCs, in turn, enhance liver metastasis of CRC cells through the TNFSF13/TNFRSF13B axis. Our study reveals the role of CRC-secreted exosomal miR-1246 in triggering HSC activation and reprogramming the TME, ultimately facilitating liver metastasis in CRC patients. Exosomal miR-1246 could serve as a potential non-invasive biomarker for predicting colorectal cancer liver metastasis, enhancing our understanding of CRC-associated liver metastases.
Collapse
Affiliation(s)
- Xiaolong Liu
- Department of General Surgery, The Third Affiliated Hospital of Southern Medical University, 183 West Zhongshan Avenue, Guangzhou, Guangdong, 510630, People's Republic of China
| | - Jialong Liu
- Department of General Surgery, The Third Affiliated Hospital of Southern Medical University, 183 West Zhongshan Avenue, Guangzhou, Guangdong, 510630, People's Republic of China
| | - Xuanyin Wang
- Department of General Surgery, The Third Affiliated Hospital of Southern Medical University, 183 West Zhongshan Avenue, Guangzhou, Guangdong, 510630, People's Republic of China
| | - Yang Zou
- Department of General Surgery, The Third Affiliated Hospital of Southern Medical University, 183 West Zhongshan Avenue, Guangzhou, Guangdong, 510630, People's Republic of China
| | - Xinyi Tao
- Department of General Surgery, The Third Affiliated Hospital of Southern Medical University, 183 West Zhongshan Avenue, Guangzhou, Guangdong, 510630, People's Republic of China
| | - Jingyu Li
- Department of Pathology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, No.106, Zhongshan 2 Road, Guangzhou, Guangdong, 510080, People's Republic of China
| | - Mengnan Ye
- Department of Pathology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, No.106, Zhongshan 2 Road, Guangzhou, Guangdong, 510080, People's Republic of China
| | - Wanbei Xu
- Department of General Surgery, The Third Affiliated Hospital of Southern Medical University, 183 West Zhongshan Avenue, Guangzhou, Guangdong, 510630, People's Republic of China
| | - Yunyao Deng
- Department of General Surgery, The Third Affiliated Hospital of Southern Medical University, 183 West Zhongshan Avenue, Guangzhou, Guangdong, 510630, People's Republic of China
| | - Lixin Liu
- Department of General Surgery, The Third Affiliated Hospital of Southern Medical University, 183 West Zhongshan Avenue, Guangzhou, Guangdong, 510630, People's Republic of China.
| | - Jingbo Sun
- Department of General Surgery, The Third Affiliated Hospital of Southern Medical University, 183 West Zhongshan Avenue, Guangzhou, Guangdong, 510630, People's Republic of China.
| | - Qingling Zhang
- Department of Pathology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, No.106, Zhongshan 2 Road, Guangzhou, Guangdong, 510080, People's Republic of China.
| |
Collapse
|
47
|
Wang Z, Ren Y, Li Y, Zhang Y, Bai S, Hou W, Zhang W, Yao Y, Zhao H, Wang M, Luo Y, Pang G, Du J. MiR-186-5p carried by M2 macrophage-derived exosomes downregulates TRPP2 expression in airway smooth muscle to alleviate asthma progression. Int Immunopharmacol 2025; 148:114107. [PMID: 39884080 DOI: 10.1016/j.intimp.2025.114107] [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: 11/12/2024] [Revised: 01/15/2025] [Accepted: 01/15/2025] [Indexed: 02/01/2025]
Abstract
Bronchial asthma (asthma) is a chronic inflammatory disease of the airways that remains an unresolved problem. Reportedly M2 macrophages and exosomes play a role in inflammation, including asthma. We investigated the roles of M2 macrophage-derived exosomes (M2-Exos) effect in asthmatic progression by using ovalbumin (OVA) induced asthmatic mice model. M2-Exos significantly ameliorated the pulmonary inflammatory response and airway hyperresponsiveness in asthmatic mice and suppressed aberrant proliferation and transient receptor potential polycystic protein 2(TRPP2) expression in LPS-stimulated primary airway smooth muscle cells (ASMCs). Then, we found that miR-186-5p of M2-Exos could target TRPP2 through online database analysis. However, miR-186-5p downregulation by miR-186-5p inhibitors decreased the protective effect of M2-Exos in asthmatic mouse and cellular models. miR-186-5p was identified and selectively combined with the polycystin-2 gene encoding TRPP2 protein, inhibited TRPP2 protein production, and downregulated TRPP2 expression. A reduction in the number of TRPP2 calcium (Ca) channels formed on the cell membrane leads to a decreased intracellular Ca2+ concentration ([Ca2+] i), causing reduced ASMC contraction and proliferation, thereby improving airway hyperresponsiveness and airway remodeling in asthma. Collectively, we conclude that M2 exosomal miR-186-5p to alleviate asthma progression and airway hyperresponsiveness though downregulating TRPP2 expression. These results may offer a novel insight to the treatment and drug delivery of asthma.
Collapse
Affiliation(s)
- Zunyun Wang
- Ciechanover Institute of Precision and Regenerative Medicine, School of Medicine, The Chinese University of Hong Kong Shenzhen Guangdong China
| | - Yan Ren
- School of Basic Medical Sciences Anhui Medical University Hefei Anhui China; The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong Shenzhen & Longgang District People's Hospital of Shenzhen Shenzhen Guangdong China
| | - Yicong Li
- School of Basic Medical Sciences Anhui Medical University Hefei Anhui China; The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong Shenzhen & Longgang District People's Hospital of Shenzhen Shenzhen Guangdong China
| | - Yuxin Zhang
- School of Basic Medical Sciences Anhui Medical University Hefei Anhui China; The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong Shenzhen & Longgang District People's Hospital of Shenzhen Shenzhen Guangdong China
| | - Suwen Bai
- The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong Shenzhen & Longgang District People's Hospital of Shenzhen Shenzhen Guangdong China
| | - Wenxuan Hou
- School of Basic Medical Sciences Anhui Medical University Hefei Anhui China; The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong Shenzhen & Longgang District People's Hospital of Shenzhen Shenzhen Guangdong China
| | - Wenjun Zhang
- School of Basic Medical Sciences Anhui Medical University Hefei Anhui China; The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong Shenzhen & Longgang District People's Hospital of Shenzhen Shenzhen Guangdong China
| | - Yanheng Yao
- School of Basic Medical Sciences Anhui Medical University Hefei Anhui China; The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong Shenzhen & Longgang District People's Hospital of Shenzhen Shenzhen Guangdong China
| | - Hongxian Zhao
- Ciechanover Institute of Precision and Regenerative Medicine, School of Medicine, The Chinese University of Hong Kong Shenzhen Guangdong China
| | - Minghua Wang
- The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong Shenzhen & Longgang District People's Hospital of Shenzhen Shenzhen Guangdong China
| | - Yumei Luo
- The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong Shenzhen & Longgang District People's Hospital of Shenzhen Shenzhen Guangdong China
| | - Gang Pang
- School of Basic Medical Sciences Anhui Medical University Hefei Anhui China.
| | - Juan Du
- Ciechanover Institute of Precision and Regenerative Medicine, School of Medicine, The Chinese University of Hong Kong Shenzhen Guangdong China; The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong Shenzhen & Longgang District People's Hospital of Shenzhen Shenzhen Guangdong China.
| |
Collapse
|
48
|
Gu Y, Mi Y, Cao Y, Yu K, Zhang Z, Lian P, Li D, Qin J, Zhao S. The lncRNA MIR181A1HG in extracellular vesicles derived from highly metastatic colorectal cancer cells promotes liver metastasis by remodeling the extracellular matrix and recruiting myeloid-derived suppressor cells. Cell Biosci 2025; 15:23. [PMID: 39972363 PMCID: PMC11841002 DOI: 10.1186/s13578-025-01365-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 02/10/2025] [Indexed: 02/21/2025] Open
Abstract
BACKGROUND Colorectal liver metastasis (CRLM) is the main cause of death in colorectal cancer (CRC) patients worldwide. In the initial stage of metastasis, primary tumors provide the necessary conditions for metastasis by shaping the local microenvironment of the target organ, forming "premetastatic niches" (PMNs), and extracellular vesicles (EVs) play important roles in shaping PMNs. Therefore, investigating the EVs involved in the regulation of PMNs and their mechanism is highly valuable for the further understanding of CRLM. METHODS Transmission electron microscopy and differential ultracentrifugation were used to verify the existence of exosomes. In vivo and in vitro assays were used to identify the roles of MIR181A1HG in EVs in CRLM. RNA pull-down and dual-luciferase reporter assays were used to clarify the mechanism by which MIR181A1HG in EVs regulated the crosstalk between CRC cells and hepatic stellate cells (HSCs). RESULTS We demonstrated that the lncRNA MIR181A1HG was progressively upregulated in tissues, serum EVs from healthy normal controls to CRC and paired liver metastatic groups. Additionally, we verified that HNRNPA2B1 mediated the packaging of MIR181A1HG into CRC cell-derived EVs, which in turn functioned as a ceRNA by sponging miR373-3p to activate HSCs via the TGFβRII/Smad2/3 signaling pathway. Furthermore, activated HSCs could secrete the chemokine CXCL12 to promote CRLM by remodeling the extracellular matrix and recruiting myeloid-derived suppressor cells in the liver, which resulted in liver metastasis. CONCLUSIONS MIR181A1HG in EVs from highly metastatic CRC cells promoted CRLM by activating HSCs to form PMNs in the liver, which contributes to the further understanding of the mechanism of CRLM and provides potential predictive markers for CRLM.
Collapse
Affiliation(s)
- Yichao Gu
- Department of General Surgery, Zhongshan Hospital, 180 Fenglin Road, Shanghai, 200032, China
| | - Yushuai Mi
- Department of Gastrointestinal Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, No. 247 Beiyuan Street, Jinan, Shandong, 250033, China
| | - Yifan Cao
- Department of General Surgery, Zhongshan Hospital, 180 Fenglin Road, Shanghai, 200032, China
| | - Kuan Yu
- Department of General Surgery, Zhongshan Hospital, 180 Fenglin Road, Shanghai, 200032, China
| | - Zihao Zhang
- Department of General Surgery, Zhongshan Hospital, 180 Fenglin Road, Shanghai, 200032, China
| | - Peng Lian
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong'an Road, Shanghai, 200032, China
| | - Dawei Li
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong'an Road, Shanghai, 200032, China.
| | - Jing Qin
- Department of General Surgery, Zhongshan Hospital, 180 Fenglin Road, Shanghai, 200032, China.
| | - Senlin Zhao
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong'an Road, Shanghai, 200032, China.
| |
Collapse
|
49
|
Zhao Y, Yu H, Li J, Qian J, Li M, Zhang X, Wang M, Wang Y, Dong Y, You Y, Zhou Q, Gao D, Zhao Y, Liu B, Chen R, Ren Z, Wang Z, Zhang K, Cui J. A glucose-enriched lung pre-metastatic niche triggered by matrix stiffness-tuned exosomal miRNAs in hepatocellular carcinoma. Nat Commun 2025; 16:1736. [PMID: 39966385 PMCID: PMC11836368 DOI: 10.1038/s41467-025-56878-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 02/03/2025] [Indexed: 02/20/2025] Open
Abstract
Apart from the classic features, it is almost unknown whether there exist other new pathological features during pre-metastatic niche formation in hepatocellular carcinoma (HCC). Our previous works have highlighted the contribution of increased matrix stiffness to lung pre-metastatic niche formation and metastasis in HCC. However, whether increased matrix stiffness influences glucose metabolism and supply of lung pre-metastatic niche remains largely unclear. Here we uncover the underlying mechanism by which matrix stiffness-tuned exosomal miRNAs as the major contributor modulate glucose enrichment during lung pre-metastatic niche formation through decreasing the glucose uptake and consumption of lung fibroblasts and increasing angiogenesis and vascular permeability. Our findings suggest that glucose enrichment, a new characteristic of the lung pre-metastatic niche triggered by matrix stiffness-tuned exosomal miRNAs, is essential for the colonization and survival of metastatic tumor cells, as well as subsequent metastatic foci growth.
Collapse
Affiliation(s)
- Yingying Zhao
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, PR China
| | - Hongmei Yu
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, PR China
| | - Jiajun Li
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, PR China
| | - Jiali Qian
- Department of Endocrinology, Huashan Hospital, Fudan University, Shanghai, PR China
| | - Miao Li
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, PR China
| | - Xi Zhang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, PR China
| | - Mimi Wang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, PR China
| | - Yaohui Wang
- Department of Interventional Radiology, Fudan University Shanghai Cancer Center, Shanghai, PR China
| | - Yinying Dong
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, Qingdao, PR China
| | - Yang You
- Department of Oncology, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Qiwen Zhou
- Department of Oncology, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Dongmei Gao
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, PR China
| | - Yan Zhao
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, PR China
| | - Binbin Liu
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, PR China
| | - Rongxin Chen
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, PR China
| | - Zhenggang Ren
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, PR China
| | - Zhiming Wang
- Department of Oncology, Zhongshan Hospital, Fudan University, Shanghai, PR China.
| | - Kezhi Zhang
- Department of Hepatobiliary Surgery, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, Jiangsu Province, PR China.
| | - Jiefeng Cui
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, PR China.
| |
Collapse
|
50
|
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] [Download PDF] [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
|